Elastin Fragments Identified as Drivers of Systemic Aging

Commentary from longevity researchers on this study:

our body’s degrading scaffold might be aging you.

The elastin holding your tissues together was mostly made in development, so damage slowly accumulates as we age, and elastin breakdown products can be detected in the blood.

Are these fragments just detritus, or do they interfere with the coordination of healthy biology?

A new paper in Nature Aging (linked below) tracked elastin fragments across a 1,068-person cohort (previous studies used ~200 people max) and found the fragments correlate with aging indicators like hypertension, BMI, liver and kidney markers, metabolic dysfunction.

To test if the fragments actually cause aging rather than just correlate with it, they gave mice elastin fragments over 24 weeks. High doses reduced lifespan by up to 17%. What’s interesting is they used much lower doses than previous studies but gave them for longer to more closely model the aging process.

The mechanism is: elastin breakdown products activate monocytes and macrophages through NEU1, which then activate T cells and drive chronic inflammation. Your body basically treats its own degraded scaffolding as a threat.

What’s interesting is they used much lower doses than previous studies but gave them for longer (24 weeks) to actually mimic the aging process rather than just acute disease. And it’s not just elastin - other ECM fragments like hyaluronan and fibronectin also shortened lifespan (though not as much).

They tested “DANA” (2,3-dehydro-2-deoxy-N-acetylneuraminic acid), a NEU1-selective sialidase inhibitor, in naturally aging mice and it extended both healthspan and lifespan (caveat, their controls were a bit short lived. Needs replication). It worked synergistically with rapamycin. DANA has been used as the structural basis for FDA-approved drugs like Tamiflu, so it’s not a brand new molecule. It has regulatory precedence.

Degraded ECM does more than simply degrade with aging. There’s evidence it also drives it, which opens up new potential anti-aging targets worth exploring.

Source: https://x.com/NornGroup/status/1994175033661333835?s=20

3 Likes

The obvious question… are there any existing drugs that inhibit NEU1? Unfortunately not.

NEU1-specific drug candidates (none approved yet)

Several groups are actively designing mammalian NEU1-selective inhibitors because NEU1 is implicated in:

  • pulmonary fibrosis
  • non-alcoholic steatohepatitis
  • cardiovascular remodeling
  • tumor progression
  • inflammatory diseases
  • ECM degradation (as in the aging study)

Promising directions:

4.1 Bicyclic and tricyclic sialic-acid analogues (2022–2024)

  • Modifications to improve NEU1 selectivity.
  • Good in vitro potency (low micromolar).
  • No in vivo data yet.

4.2 Macrocyclic NEU1 inhibitors

  • Bind a secondary pocket unique to NEU1.
  • Currently only in enzyme assays.

4.3 Covalent NEU1-targeted ligands

  • Designed using fragment-based screening.
  • Possibly the most promising, but pre-preclinical.

4.4 NEU1 antibodies / biologics

  • In development but NEU1 is largely intracellular/lysosomal, making targeting difficult.

None are anywhere near FDA approval.


5. Bottom Line

There are currently zero approved drugs that meaningfully inhibit NEU1.

The only compounds showing real inhibition are:

  • DANA (weak, broad, not drug-like)
  • DANA derivatives (experimental)
  • Novel inhibitors from 2022–2024 medicinal chemistry literature
  • RNA-based methods (laboratory use only)

Some approved drugs can modulate NEU1 expression , or indirectly reduce its functional signaling , but none inhibit its enzymatic activity .

For longevity science:

The NEU1 pathway is druggable, but currently not drugged. The mouse lifespan-extension result will almost certainly catalyze development of NEU1-selective therapeutics, much like how DANA led to modern antivirals.

2 Likes

Isn’t this hyaluronic acid? This is commonly used in skincare and also is a supplement, which I take. I need to look more into this. GPT theorized that supplementing isn’t the same.

Also this entire thing is very interesting. We are getting close to cleaning up the mess of elastin breakdown, but does this give us any insight into preventing elastin breakdown, or slowing it, or reversing it?

Elastinase inhibitors? Though it seems that might prevent the breakdown of fragments.

I ran all of this post through ChatGPT then tried to figure out what elastinase inhibitors already exist and utilizing what is currently approved, what is in clinical trials, research chemicals and more what would be the most aggressive combination we could use to slow down or reverse damage to elastin:

Let’s put together a hypothetical, maximalist ECM/elastin preservation protocol based on everything known today—approved drugs, off-label uses, research chemicals, natural compounds, and lifestyle interventions. I’ll structure it in mechanistic layers, from upstream prevention of breakdown to downstream modulation of NEU1 signaling and systemic inflammation. This is purely theoretical and not a prescription.


1. Upstream: Reduce Elastin Degradation

Goal: Minimize elastase activity and ECM breakdown.

Pharmacologic/Research Options:

  • MMP inhibitors (experimental/research use):
    • Broad-spectrum MMP inhibitors exist (batimastat, marimastat) but toxic long-term. Consider low-dose selective MMP inhibitors if available in preclinical settings (MMP-9, MMP-12 inhibitors).
  • Serine elastase inhibitors:
    • Sivelestat – inhibits neutrophil elastase (studied for ARDS). Could theoretically reduce systemic elastin breakdown from inflammation.
  • Cathepsin inhibitors:
    • Odanacatib (cathepsin K inhibitor) reduces elastin and collagen degradation in bone/vascular tissue.
  • Senolytics:
    • Dasatinib + Quercetin – reduce senescent cells, the major source of elastase activity.
    • Fisetin is a milder alternative, may be easier for repeated late-life dosing.

Natural Compounds:

  • EGCG (green tea extract) – inhibits MMPs, antioxidant.
  • Curcumin / Piperine combo – MMP suppression and anti-inflammatory.
  • Quercetin – overlapping senolytic and MMP inhibition.

Lifestyle Measures:

  • UV avoidance / sunscreen to prevent photoaging of elastin.
  • Blood pressure control (hypertension accelerates elastin damage in arteries).
  • Anti-inflammatory diet (omega-3s, polyphenols, low glycemic load).
  • Exercise to maintain healthy tissue turnover.

2. Midstream: Neutralize Elastin Fragments / NEU1 Signaling

Goal: Stop fragments from triggering inflammation.

Experimental Compounds:

  • DANA (2,3-dehydro-2-deoxy-N-acetylneuraminic acid) – weak NEU1 inhibitor, proof-of-concept. Can combine with rapamycin for synergy.
  • Rapamycin / mTOR inhibitors – suppress downstream nutrient-sensing-driven aging; synergistic with NEU1 inhibition.
  • Novel NEU1-selective inhibitors – if accessible in research settings, these are the most direct intervention.

Indirect Modulation:

  • Some approved drugs or nutraceuticals may downregulate NEU1 expression indirectly (less well-studied).

3. Downstream: Reduce Systemic Inflammation & ECM Remodeling

Goal: Prevent T-cell mediated inflammaging and organ dysfunction.

Pharmacologic Options:

  • Senolytics – continue periodic dosing (D + Q, Fisetin, or Navitoclax derivatives) to prevent ongoing elastin release.
  • Anti-inflammatories:
    • Low-dose metformin (AMPK activation, anti-inflammatory).
    • IL-1β inhibitors (canakinumab – research use for inflammaging).
  • Immune modulators:
    • Rapamycin, as above.
    • Possibly thymic rejuvenation approaches (IL-7, growth hormone + thymosin alpha1) to maintain healthy adaptive immunity.

Lifestyle Measures:

  • Caloric/nutrient optimization to reduce metabolic stress.
  • Structured exercise to maintain tissue elasticity and vascular health.

4. Optional: ECM Repair / Regeneration

Goal: Stimulate new elastin or prevent crosslinking degradation.

Experimental Approaches:

  • LOX inhibition (lysyl oxidase modulators) – prevent pathological crosslinking in arteries and skin.
  • TGF-β pathway modulation – can enhance elastin repair but risky if overdone (fibrosis risk).
  • Gene therapy / peptide approaches:
    • Elastin mimetic peptides (like VGVAPG analogues in reverse) or micro-elastin constructs – preclinical only.
  • Stem cell or regenerative therapies – mesenchymal stem cells have been shown to support ECM maintenance in aged tissues (mostly in preclinical or clinical trials).

5. Hypothetical Integrated Protocol

Daily/Weekly Actions:

  1. Senolytic pulse – Fisetin or D + Q once every few weeks/months.
  2. Rapamycin / mTOR modulation – weekly low-dose dosing.
  3. NEU1 modulation – DANA or research NEU1 inhibitor if available.
  4. Anti-elastase support – EGCG, curcumin, quercetin daily. Optional research elastase inhibitors.
  5. Lifestyle – strict UV avoidance, BP control, exercise, anti-inflammatory diet.

Monitoring (research settings):

  • Circulating elastin fragment levels.
  • Inflammatory cytokines (IL-1, IL-6, TNF-α).
  • Body composition, grip strength, and metabolic parameters.

In short, the ultimate approach would be multi-layered: reduce elastin breakdown (senolytics + elastase inhibitors), block fragment signaling (NEU1 inhibition), modulate immune response (rapamycin, anti-inflammatories), and support ECM repair where possible.

2 Likes

Its all about the fragment size it seems, and the systemic circulation (not an issue with topicals):

How systemic HA affects longevity

  • Reduces inflammation: High-molecular-weight HA can directly suppress the production of pro-inflammatory factors by immune cells. It can also form physical structures that prevent inflammatory cells from activating, according to this National Institutes of Health (NIH) study.
  • Combats oxidative stress: HA has the ability to scavenge reactive oxygen species, which are damaging byproducts of metabolism that contribute to aging. This protective function is particularly important for long-lived species.
  • Maintains tissue integrity: By supporting the health of the intestinal stem cells, HA helps prevent age-related decline in the intestinal barrier, which is crucial for overall health.
  • Decreases cancer risk: Research indicates that increased HA levels are associated with a lower incidence of both spontaneous and induced cancers in mice, notes this National Institutes of Health (.gov) article.
  • Improves healthspan: By mitigating age-related issues like inflammation and oxidative stress, systemic HA contributes to a longer and healthier life, not just a longer one.

Key distinction: HA size matters

  • The beneficial effects are primarily linked to high-molecular-weight (HMW) HA .
  • Fragmented or low-molecular-weight HA, often produced during inflammation, can have pro-inflammatory effects.
  • The specific effects of HA are complex and depend on factors like size, concentration, and microenvironment, says this NIH article.
2 Likes

Focusing just on this, I dug in deeper. But we also really need to dig deep into the side effects and regular applications of these medications. This is just a start of the learning process.

Prompt: Can you identify the relative effect sizes of these interventions listed here, in terms of their ability to Reduce Elastin Degradation

Response:

Below is a direct, evidence-weighted ranking of the interventions you listed, organized by estimated effect size on elastin preservation (i.e., inhibition of elastases, MMPs, or cathepsins; reduction of ECM-degrading inflammatory programs; or lowering of senescent-cell–derived proteases).

+++ large effect (direct elastase/MMP/cathepsin inhibition; strong senolytic reduction of NE/MMP-rich SASP)

++ moderate effect (potent but indirect modulation; requires high doses or specific delivery)

+ small effect (nutraceutical-level, often weak vs. pharmacologic agents)


Relative Effect Size Ranking (Summary)

Rank Intervention Estimated Effect Size Mechanism
1 Sivelestat +++ Direct neutrophil elastase inhibition
1 Odanacatib +++ Direct cathepsin K inhibition (major elastase)
1 Dasatinib + Quercetin +++ Senescent-cell removal → large drop in elastases/MMPs
2 Selective MMP inhibitors (MMP-9, MMP-12) ++ to +++ Blocks key elastin-degrading MMPs
2 Fisetin ++ Partial senolytic; SASP suppression
3 EGCG + to ++ Mild MMP inhibition
3 Curcumin + Piperine + Weak indirect MMP/NE suppression
3 Quercetin alone + Mild antioxidant/MMP inhibition

Interpretation & Confidence

Best overall strategy for minimizing elastin degradation:

  1. Neutralize the major proteases (NE, CatK, MMP-12)
  2. Reduce the cellular sources (senescent cells, activated neutrophils)
  3. Maintain low systemic inflammation (IL-6, TNF-α)

Confidence Levels

  • High for sivelestat, odanacatib, D+Q (strong mechanistic and preclinical evidence).
  • Moderate for MMP inhibitors (target-specific, toxicity concerns).
  • Low–moderate for nutraceuticals (effect sizes small; dosing constraints).

Full response here:

1 Like

I feel like cycling through some of these and intermittently using some like we do Rapamycin would be the best thing. Almost certainly the things being inhibited exist for a reason and have some benefit so by only inhibiting a few things at a time and cycling through them we would get maximal benefit.

We need more studies though… I wish I was a billionaire haha.

1 Like

They are interesting… pulsed dosing is one strategy that would seem potentially effective while minimizing side effect risks. Datatinib is the only one of these drugs available from India (Sivelestat (marketed as “Elaspo”) seems to be hard to get (not available from India), and an injection drug, Odanacatib was canceled during development).

But - when Sivelestat was launched it was priced at about $40 per 100mg vial, which as far as new drug pricing goes seems amazingly inexpensive (seems the Japanese drug companies are a little less mercenary than the US/Europeans). So if you can get it, it’s probably not cost-prohibitive.

Below are concise, side-effect–focused profiles for each compound, emphasizing frequency of clinically significant adverse effects based on trial data, FDA filings (where applicable), and major published studies. Embedded links give primary literature or regulatory sources.


Sivelestat

Type: Neutrophil elastase inhibitor (IV), approved only in Japan/South Korea for ARDS.
Key safety profile: Generally well tolerated; adverse-event rates modest but include meaningful hepatic and hematologic signals.

Significant side effects (approx. frequency ranges):

  • Liver enzyme elevation: ~5–15% in clinical studies
  • Thrombocytopenia: ~2–5%
  • Renal dysfunction: ~2–4%
  • GI symptoms (nausea, diarrhea): ~5–10%
  • Serious adverse events overall: Not significantly higher than placebo in ARDS trials

Special note: No chronic use safety data; all studies involve short-duration infusion in critical illness.


Odanacatib

Type: Cathepsin K inhibitor for osteoporosis (development terminated).
Key safety profile: Highly effective anti-resorptive drug but major issue was increased stroke risk → program halted by Merck.

Special note: Drug was otherwise well tolerated; the stroke signal was the decisive safety failure.


Dasatinib

Type: Multi-kinase inhibitor (SRC/ABL), FDA-approved for CML; used in low-dose senolytic regimens experimentally.
Key safety profile: Well characterized; significant cytopenias and pleural effusion at standard oncology doses.
Significant side effects (standard 100 mg/day dosing):

  • Myelosuppression:
    • Neutropenia: ~20–35%
    • Thrombocytopenia: ~15–30%
    • Anemia: ~10–15%
      (FDA label)
  • Pleural effusion: ~15–30% (dose- and duration-dependent)
  • Fluid retention (general): ~10–20%
  • QT prolongation: <1%
  • GI toxicity (diarrhea, nausea): 10–20%
  • Infection risk: Increased due to cytopenias (~10–20%)

Low-dose senolytic use (e.g., 2–10 mg):

  • Limited human data; adverse events appear rare (<5%) in short-pulse regimens, but datasets remain small and heterogeneous.

1 Like

Just testing out this research tool that @AlexKChen pointed out:

A Stanford / Berkeley AI Research Tool focused on academic papers (bioRxiv, etc.)

The results for my search on NEU1 inhibitors:

NEU1inhibitors_research-report.pdf (1.2 MB)

1 Like

Elastin_research-report.pdf (1.2 MB)

1 Like

Gemini 3.0 Pro: Based on the mechanisms identified in the paper—specifically the Elastin-NEU1-Immune axis —here are 10 suggestions for natural interventions and off-label drugs that could theoretically interrupt this pathway.

These suggestions target three specific points in the pathway described in the study:

  1. Inhibiting NEU1 (the receptor that triggers the immune response).

  2. Inhibiting Elastase (MMP-12/NE) (the enzymes that chop elastin into toxic fragments).

  3. Clearing Senescent Cells (which the paper identified as the source of the elastase).


Off-Label Pharmaceutical Drugs

1. Oseltamivir (Tamiflu)

  • Mechanism: Direct NEU1 Inhibition.

  • Why it fits: The paper demonstrates that DANA is the molecule that extends lifespan by inhibiting NEU1. The discussion section explicitly notes that DANA shares the “parent scaffold” of FDA-approved neuraminidase inhibitors like Oseltamivir. While Oseltamivir is designed for viral neuraminidase, it has cross-reactivity with human mammalian neuraminidase 1 (NEU1).

  • Target: Blocks the receptor (NEU1) from recognizing the elastin fragment (E-motif).

2. Rapamycin (Sirolimus)

  • Mechanism: mTOR Inhibition & Synergy.

  • Why it fits: The paper explicitly tested Rapamycin in combination with DANA. The results showed that Rapamycin + NEU1 inhibition had a synergistic effect, extending lifespan and healthspan significantly more than either treatment alone. Rapamycin also reduces the secretion of inflammatory factors (SASP) that degrade the extracellular matrix.

  • Target: Downstream inflammation and upstream cellular senescence.

3. Metformin

  • Mechanism: Anti-Glycation & ECM Protection.

  • Why it fits: Elastin degradation is accelerated by glycation (the formation of Advanced Glycation End-products, or AGEs), which makes elastin brittle and prone to fragmentation. By improving insulin sensitivity and reducing blood glucose, Metformin prevents the structural damage that leads to the creation of the toxic fragments described in the paper.

  • Target: Prevents the formation of elastin fragments.

4. Doxycycline (Low Dose)

  • Mechanism: MMP Inhibition.

  • Why it fits: The paper identifies Macrophage Elastase (MMP-12) as a primary enzyme responsible for generating toxic elastin fragments. Doxycycline is a well-known, potent inhibitor of Matrix Metalloproteinases (MMPs), widely used off-label in dermatology to prevent collagen and elastin destruction.

  • Target: Inhibits the enzymes (MMPs) that cut elastin into toxic fragments.


Natural Interventions & Supplements

5. Quercetin (specifically with a Senolytic agent)

  • Mechanism: Senolytic Activity.

  • Why it fits: The study explicitly performed an experiment using Dasatinib + Quercetin (D+Q) . They found that clearing senescent cells significantly reduced the serum levels of elastin fragments. The researchers concluded that senescent cells are the “factory” producing the elastases that degrade the matrix.

  • Target: Eliminates the cells producing the elastin-degrading enzymes.

6. EGCG (Green Tea Extract)

  • Mechanism: Elastase Inhibition.

  • Why it fits: Epigallocatechin gallate (EGCG) has been shown in multiple studies to inhibit Macrophage Elastase (MMP-12) and Neutrophil Elastase , the exact two enzymes the paper identifies as responsible for cleaving elastin. By inhibiting these enzymes, fewer “E-motif” fragments are released into circulation.

  • Target: Direct inhibition of elastin degradation.

7. Boswellia Serrata (Frankincense)

  • Mechanism: Neutrophil Elastase Inhibition.

  • Why it fits: The paper identifies Neutrophil Elastase (NE) as a key driver of elastin fragmentation. Boswellic acids are among the few known natural, specific, non-competitive inhibitors of human leukocyte elastase (HLE/Neutrophil Elastase).

  • Target: Stops neutrophils from breaking down elastin.

8. Grape Seed Extract (Procyanidins)

  • Mechanism: Elastin Stabilization.

  • Why it fits: Procyanidins promote the cross-linking of elastin fibers, making them more resistant to enzymatic hydrolysis (being chopped up). If the elastin is stabilized, it cannot be broken down into the “VGVAPG” oligopeptides that trigger the NEU1 receptor.

  • Target: Hardens the extracellular matrix against degradation.

9. Curcumin (Turmeric)

  • Mechanism: NF-κB Inhibition.

  • Why it fits: The paper notes that when the E-motif binds to the receptor, it activates the NF-κB pathway inside monocytes and macrophages, leading to inflammation (IL-1, IL-6). Curcumin is a potent NF-κB inhibitor, theoretically blocking the “signal” even if the elastin fragment binds to the cell.

  • Target: Blocks the downstream inflammatory signal inside the immune cell.

10. Gotu Kola (Centella Asiatica)

  • Mechanism: ECM Remodeling & Synthesis.

  • Why it fits: While the paper focuses on degradation, the antidote is repair. Centella Asiatica is one of the few herbs proven to stimulate fibroblast proliferation and the synthesis of new collagen and fibronectin, helping to maintain the integrity of the extracellular matrix before it degrades into toxic matrikines.

  • Target: ECM maintenance and repair.

4 Likes

I’ve called this document “Mechanistic Framework for Aortic Root Shrinkage”, something that has not been achieved yet… maybe.
0qg1wv.pdf (127.1 KB)

It’s suppositional, due to the dill/blackberry extracts, but I am adding it because it also discusses preservation of the ECM. I also added brand names in the workup, just for simplicity (and ranking?).

2 Likes

Some follow-on questions on the original research paper summarized:

I’ll go through the questions one by one and separate what’s known, what’s plausible, and what’s still guesswork. (Editor: I’ve eliminated some of the questions that were not very relevant here)


1. Can we measure elastin fragments (ELN fragments) in humans today?

Yes, but only as research assays, not standard clinical tests.

  • Multiple companies sell human elastin-derived peptide (EDP) ELISA kits for serum/plasma (e.g., “Human EDP (Elastin-Derived Peptides) ELISA Kit” and similar). These are marketed strictly for research use, not for clinical decision-making.
  • There are also ELISAs for total elastin in serum/plasma.
  • In principle, targeted LC–MS/MS proteomics can quantify specific elastin peptides, and this is in line with what the Nature Aging paper did for murine/human cohorts (though their exact assays are not commercially packaged).

Gaps:

  • No standardized reference ranges by age/sex.
  • No CLIA-validated test you can order from a mainstream lab yet.
  • No longitudinal human data on “track EDP every 6–12 months and correlate with outcomes.”

For a biohacker: feasible only via research collaboration or paying for custom proteomics / RUO ELISAs; not plug-and-play like hs-CRP.


2. Can lifestyle/environment reduce elastin fragmentation in humans?

We have strong indirect evidence; very limited direct EDP data.

Most of the following are almost certainly elastin-sparing, even if the exact EDP delta isn’t quantified:

  1. Smoking / inhaled toxins
  • Smoking drives elastase-mediated destruction of lung elastin in COPD/emphysema and increases elastase in the circulation. Smoking cessation consistently slows radiographic and functional decline in lungs, which is mediated in part via lower elastase activity.
  1. Blood pressure and vascular stress
  • Chronic hypertension accelerates arterial elastin fatigue and fragmentation, contributing to arterial stiffness and aneurysm formation. Tight BP control reduces progression of arterial stiffness and aortic dilation in multiple cohorts.
  1. UV exposure
  • Photoaging literature is very clear that chronic UV causes solar elastosis and dermal elastin breakdown; UV-protected vs sun-exposed skin shows striking elastin degradation and abnormal elastin aggregates histologically.
  1. Chronic lung disease control (COPD, bronchiectasis, severe asthma)
  • These states are rich in neutrophil elastase. Neutrophil elastase inhibitors like sivelestat are used acutely in ARDS/ALI in Japan/China, precisely because NE drives lung injury.
  1. Oral and systemic inflammatory burden
  • Periodontal disease, obesity, and metabolic syndrome all increase neutrophil and monocyte activation. They plausibly raise elastase activity and ECM turnover, though this is mostly extrapolated from inflammatory and MMP data not direct EDP measurements.

Take-home: if your goal is to minimize elastin fragmentation, the usual longevity hygiene (no smoking, low BP, low visceral fat, low chronic inflammatory load, UV protection, aggressive periodontal care) is highly coherent with the mechanism, even if we don’t yet have human EDP curves pre- and post-intervention.


4. Existing drugs that reduce elastase activity without insane risk?

Here we’re mainly talking about neutrophil elastase (NE) and related proteases .

4.1 Sivelestat

  • A selective neutrophil elastase inhibitor used in Japan and parts of Asia for acute lung injury/ARDS, usually IV and short-term.
  • Meta-analyses suggest modest benefit in ARDS; evidence in chronic respiratory disease (COPD, CF, bronchiectasis) is weak or negative.
  • No long-term safety data for chronic use; no evidence for aging or ECM endpoints.

This is not a very practical longevity drug for most people. It’s approved only in Japan / Asia, and is a hospital-grade acute care drug with IV delivery. Hard to get, though cost is low because it seems to have gone generic (drug was introduced in 2002).

Grok: Sivelestat (also known as Elaspol) is not sold by any online pharmacies. It is a prescription medication approved only in Japan and South Korea.

Sivelestat (known as 西维来司他钠 or Elaspol in Chinese) is sold in China. It was approved by the National Medical Products Administration (NMPA) in March 2020 via an expedited emergency review process for treating acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) associated with systemic inflammatory response syndrome (SIRS), particularly during the COVID-19 pandemic. Developed by Shanghai Huilun Jiangsu Pharmaceutical Co., Ltd., it has been widely used in hospitals, with over 1.9 million units distributed by 2022 for ICU and respiratory care. As of 2024, it remains a top-selling respiratory drug in public hospitals and is included in national procurement programs. It is available through hospital pharmacies and authorized medical channels, not over-the-counter or general retail.

Details on China Manufacturer and Product: 注射用西维来司他钠 - 上海汇伦医药股份有限公司

4.2 Doxycycline and other tetracyclines

  • Doxycycline at sub-antimicrobial doses is used in periodontal disease and some aneurysm/MMP-driven conditions; it inhibits MMPs more than neutrophil elastase, but net effect is ECM-protective.
  • Chronic use has tolerability and microbiome issues; again, no hard data on systemic EDP.

4.3 Inhaled or local elastase inhibitors

  • Various inhaled NE inhibitors have been trialed in CF/COPD; thus far, no robust clinical success.

4.4 Realistic longevity stance

Right now, no approved drug is suitable for chronic systemic NE inhibition as a longevity strategy.


5. Nutraceuticals / polyphenols / peptides that modulate NEU1 or the elastin receptor?

5.1 NEU1-selective inhibitors (not nutraceuticals)

  • Medicinal chemistry has produced NEU1-selective DANA derivatives, e.g. C9-butyl-amide-DANA, which blocks NEU1 activity and NEU1-mediated responses in human lung cells and mouse lung models.
  • Newer work (2022–2025) reports additional NEU1-biased sialidase inhibitors with IC₅₀ in the low micromolar range.
  • All are research compounds, not supplements or drugs.

5.2 Interfering peptides / biologics

  • NEU1-targeting peptides have been used to inhibit NEU1 activity in insulin signaling and other pathways in animals.
  • Again: purely experimental.

5.3 Polyphenols etc.

  • Some polyphenols (EGCG and others) show weak, broad glycosidase inhibition in vitro at millimolar concentrations—far above what you achieve with oral dosing. No credible evidence they meaningfully inhibit NEU1 in vivo.
  • Anything claiming to be a “NEU1 inhibitor supplement” right now is marketing, not science.

So: there are no nutraceutical-grade NEU1 inhibitors with convincing data. The real action is in DANA derivatives and NEU1-specific medicinal chemistry, all preclinical.


6. Downstream biomarkers accessible today

You can’t yet buy a NEU1 or EDP panel from Quest/Labcorp, so you lean on downstream and adjacent markers.

Reasonable proxy/read-out markers :

  • hs-CRP: standard low-grade inflammation marker; not specific, but robust and cheap.
  • GlycA (NMR glycoprotein acetyls): integrated marker of systemic inflammation and subclinical vascular inflammation, predictive of cardiometabolic risk and CVD events.
  • Neutrophil elastase activity: some research labs and niche services offer NE activity assays in plasma or BAL; not widely available clinically.
  • Monocyte/NLR metrics :
    • CBC-derived neutrophil-to-lymphocyte ratio (NLR) as a crude inflammaging marker.
    • Flow cytometry panels (research setting) for inflammatory monocyte subsets (CD14⁺⁺CD16⁺⁺, etc.) and T-cell phenotypes (CD28null, KLRG1⁺, etc.).
  • Arterial stiffness :
    • Pulse wave velocity (PWV) and central aortic pressure; elastin degradation is a major contributor.
  • Multi-omics clocks :
    • Proteomic clocks that are enriched for ECM and inflammatory proteins; the organ-specific proteomic aging clocks you’ve already looked at are conceptually compatible with this axis.

None of these are specific to ELN/NEU1, but if you’re trying to see whether a regimen is reducing chronic vascular and myeloid inflammation, they are reasonable surrogates.


8. Could intermittent senolytics (D+Q) lower elastin fragmentation in humans?

Mechanistically plausible; empirically unproven.

What we know:

  • In mice, D+Q reduced senescent myeloid cells and elastase activity and lowered elastin fragments. That’s directly in the paper.
  • In humans, small trials show that intermittent D+Q:
    • reduces senescent cell markers and SASP factors in adipose tissue and blood,
    • improves some functional measures (e.g., mobility, bone metabolism).
  • No published human data yet on EDP/ELN-fragment levels before/after senolytic treatment.

So, it is reasonable to hypothesize that effective senolysis will indirectly reduce elastase load and elastin fragmentation over time—but we don’t have measurements. Until we see actual EDP or ECM-related imaging endpoints in senolytic trials, it’s an open question.


9. At what age does ELN-fragment accumulation become relevant for people?

From the Nature Aging paper’s human cohort:

  • ELN fragments increase with chronological age across adult life and correlate with worse cardiometabolic and renal markers and higher blood pressure. Exact age cut-points in that dataset are not fully detailed in the abstract, but the pattern is monotonic.
  • Other ECM-related changes (arterial stiffness, PWV, collagen/elastin remodeling) start to diverge measurably from early to mid-adulthood, especially in people with hypertension, obesity, or smoking history.

Reasonable inference:

  • For a metabolically healthy, normotensive, non-smoker, early 20s–30s probably have low EDP load.
  • By 30s–40s, lifestyle and risk factors start to diverge, likely producing different EDP trajectories.
  • By 50+, EDP is almost certainly relevant as a systemic biomarker of vascular and organ aging.

But since there are no public EDP reference curves by age, we can’t draw a clean line like “age 35 is when you must intervene.” The rational stance: treat ECM-sparing behaviors as life-long, not late-life rescue.


10. Realistic timelines for NEU1-selective therapeutics

State of the field:

  • Recent reviews emphasize NEU1 as a target in fibrosis, cardiovascular disease, and inflammation, and summarize NEU1-selective inhibitors (e.g., C9-butyl-amide-DANA) with in vitro and in vivo lung data.
  • 2022–2025 medicinal chemistry papers report newer NEU1-biased compounds with micromolar potency, but no human PK, no toxicity, no phase I yet.

Drug-development reality:

  • From “tool compound with animal efficacy” → “approved human drug” is typically 10–15 years if everything goes smoothly (and many fail).
  • NEU1 is also lysosomal and membrane-associated, raising complexities in achieving tissue-specific, safe inhibition.

So best guess :

  • 0–5 years: more animal data (fibrosis, cardiovascular, metabolic, possibly aging) + first-in-human safety studies if a candidate looks clean.
  • 5–10+ years: if successful, early disease-specific applications (pulmonary fibrosis, NASH, maybe vascular remodeling).
  • Aging/geroprotection as a primary indication would likely be even further out, if it happens at all.

I wonder how specific UVB, as in vitamin-D lamps like Sperti, figures into this, since we need vitamin D to heal (etc), and oral isn’t the same as skin synthesis.

1 Like

One has to ask to what detriment would inhibiting Neu1 lead to…

What is NEU1?
NEU1 is a lysosomal enzyme that removes sialic acid residues from glycoproteins, glycolipids, and oligosaccharides. It plays a critical role in the catabolism of sialoglycoconjugates. Deficiency or dysfunction of NEU1 leads to accumulation of oversialylated substrates in lysosomes and altered cell signaling.

NEU1 deficiency syndromes
Severe NEU1 mutations cause sialidosis (type I and II), a rare lysosomal storage disease.
Partial or hypomorphic NEU1 deficiency is strongly linked to arterial disease, especially aortic root dilation and aneurysms.

Key vascular effects of reduced NEU1 activity

  1. Elastic fiber fragmentation and loss

    • NEU1 normally degrades oversialylated glycans on elastin and associated proteins (fibrillin-1, fibulin, etc.).
    • When NEU1 is deficient, excessive sialylation accumulates on elastic fibers → makes them stiffer and more susceptible to proteolytic degradation by matrix metalloproteinases (MMPs, especially MMP-2 and MMP-9).
    • Result: breakdown of elastic lamellae in the aortic media → medial degeneration and progressive aortic dilation.
  2. Smooth muscle cell (SMC) phenotypic switching

    • Reduced NEU1 causes hypersialylation of cell-surface receptors (e.g., PDGF receptor, EGFR, IGFR).
    • This prolongs and amplifies growth-factor signaling → SMCs switch from contractile to synthetic/proliferative phenotype → intimal thickening and further weakening of the aortic wall.
  3. Lysosomal exocytosis and cathepsin/MMP release

    • Accumulated substrates cause lysosomal stress → increased exocytosis of lysosomal contents (cathepsins, MMPs) into the extracellular space → additional elastolysis and collagen degradation.
  4. Inflammation and oxidative stress

    • NEU1-deficient cells show increased ROS and inflammatory cytokine production, further driving vascular remodeling.

Clinical and genetic evidence

  • Biallelic hypomorphic NEU1 mutations are found in a significant percentage of patients with isolated thoracic aortic aneurysms (especially early-onset or familial cases that are negative for classic connective-tissue disease genes such as FBN1, TGFBR1/2, SMAD3, ACTA2, etc.).
  • A common NEU1 variant (p.Arg341Ala, previously called p.Arg347Gln) dramatically reduces enzyme activity and is enriched in aortic aneurysm cohorts.
  • Mouse models:
    • Neu1 knockout mice die perinatally, but heterozygous Neu1+/− mice and hypomorphic Neu1V98M/V98M knock-in mice develop progressive aortic dilation, elastic fiber fragmentation, and medial degeneration that closely resembles human disease.

Human diseases linked to NEU1 and aortic pathology

  • Sialidosis (severe mutations) – rare, multisystem, sometimes with mild aortic root enlargement.
  • NEU1-related isolated thoracic aortic aneurysm and dissection (partial deficiency).
  • Some overlap with cutis laxa and arterial tortuosity syndromes when combined with other glycosylation defects.

Summary of the mechanism leading to aortic dilation

Low NEU1 activity

Hypersialylation of glycoproteins in elastic fibers and SMC receptors

Elastic fiber degradation + excessive growth-factor signaling

Medial degeneration, loss of elasticity, SMC loss or maladaptive remodeling

Progressive aortic root/ascending aortic dilation ± dissection risk

anyhow, best watch out turning that Neu1 down too much.

1 Like

As with all medications, “dose makes the poison”. The citations you mentioned are really focused around complete inhibition of NEU1, it seems. Completely shut down any significant biological pathway and you’ll have problems eventually, especially if started early in life. But later, with moderation (such as with mTOR) can have very beneficial effects.

At first it can be seen as a paradox, how could inhibition in the first paper result in life extension, but in your quotes its life shortening. But, as CGPT suggests:

The “paradox” with lifespan extension from NEU1 inhibition disappears once you distinguish (i) congenital/near-total NEU1 loss vs (ii) partial, late-life inhibition of overactive NEU1 signaling at the cell surface.

“NEU1 deficiency syndromes” and vascular disease

“Severe NEU1 mutations cause sialidosis… Partial or hypomorphic NEU1 deficiency is strongly linked to arterial disease, especially aortic root dilation and aneurysms.”

The first sentence is correct; the second is overstated and not well supported.

What we actually know:

  • Neu1-null mice (a sialidosis model) have abnormal elastic fiber assembly in aorta and lung: thinner elastic lamellae, excess sialylated moieties, altered fibrillin deposition.
  • NEU1 is required for normal elastogenesis and elastic fiber homeostasis, especially during development.
  • In humans, NEU1 mutations primarily present as classic sialidosis (neurologic, skeletal, renal, etc.). A strong, clean epidemiologic link “hypomorphic NEU1 → aortic root aneurysm” is not well documented; aneurysms and aortic disease are dominated by other genes (FBN1, TGFβ pathway, etc.) and risk factors.

Conversely, in adult vascular disease you actually see the opposite pattern:

  • Macrophage-derived NEU1 promotes aortic dissection and ECM degradation in a BAPN mouse model; myeloid-specific NEU1 knockout ameliorates vascular inflammation, ROS, apoptosis and reduces dissection .

So: NEU1 deficiency clearly causes a storage disease with abnormal elastin assembly, but “partial NEU1 deficiency → aneurysm” is at best a niche hypothesis, not a well-established fact.

“Inflammation and oxidative stress”

“NEU1-deficient cells show increased ROS and inflammatory cytokine production, further driving vascular remodeling.”

Here the picture is context-dependent and your sentence is too one-sided:

  • In many cardiac, renal, and vascular injury models, NEU1 overexpression is associated with more oxidative stress and inflammation, and NEU1 inhibition or deficiency is protective, improving cardiac function or reducing fibrosis and vascular damage.
  • In immune cells (microglia, macrophages), NEU1 activity is required for TLR and other receptor desialylation; NEU1 knockdown reduces cytokine induction in response to LPS and other stimuli.

So depending on cell type and disease context, too much NEU1 activity or too little can both be harmful, but your “NEU1 deficiency → more ROS/inflammation” is not generally true.

Practical implication for longevity thinking

Your summary text captured one important side (NEU1’s role in elastogenesis and lysosomal homeostasis), but the aging and cardiovascular literature over the last ~5–10 years has made it clear that NEU1 is also a pro-inflammatory, pro-fibrotic effector when overactive in adult tissues.

That’s why inhibiting NEU1 in old mice with pre-existing elastin damage extends lifespan, even though total NEU1 loss from birth causes a lethal storage disease.

What’s still unknown (and non-trivial):

  • The safe therapeutic window of NEU1 inhibition in long-lived mammals.
  • Whether chronic strong inhibition would, over decades, begin to reproduce aspects of sialidosis.
  • How selectively we can target cell-surface / immune-cell NEU1 vs global lysosomal NEU1.
2 Likes

This is how a N=1 clinical trial might be handled (not encouraging it, just doing this as a thought experiment to stimulate discussion).

It seems likely that Sivelestat (known as 西维来司他钠 or Elaspol in Chinese) is likely to be not too difficult to obtain in China…

Short answer: based on what’s actually published by Chinese hospitals and regulators, Sivelestat sodium injection (注射用西维来司他钠, 0.1 g vial) is effectively nationally standardized at ~249 CNY per vial, with a few hospitals slightly higher (~276 CNY). At today’s FX (≈0.141 USD/CNY), that’s roughly 35–39 USD per 0.1 g vial. Regional variation is small.

and the question becomes:

What would a reasonable but simple and cost-effective N=1 clinical trial look like for using Sivelestat to see if it can reduce systemic elastin breakdown from inflammation.

AI Response:

The following is a blueprint for a reasonable, scientifically grounded N=1 trial for Sivelestat in a healthy older male.

1. The Core Challenge: Route of Administration

Before detailing the protocol, there is a critical operational hurdle: Sivelestat is only available as an Intravenous (IV) formulation.

  • Availability: There is no oral pill.
  • Pharmacokinetics: It has a very short half-life (~2 hours).
  • Standard Use: In ICU settings (for ARDS/ALI), it is given as a 24-hour continuous infusion.

The “Simple” Solution:

You cannot reasonably keep a healthy volunteer on a continuous pump for days. However, pharmacokinetic studies in healthy volunteers have successfully used intermittent bolus infusions.

  • Proposed Protocol Adjustment: Instead of continuous infusion, use 2-hour IV infusions twice daily (every 12 hours). This mimics the dosing used in healthy volunteer safety studies (e.g., Chinese cohorts) without requiring hospital admission.

2. Clinical Protocol Design (N=1)

Objective

To determine if acute inhibition of Neutrophil Elastase (NE) with Sivelestat reduces urinary excretion of desmosine (a specific biomarker of elastin breakdown) in a healthy older male.

Participant

  • Inclusion: Healthy male, age >60 (aging is naturally associated with low-grade inflammation or “inflammaging”).
  • Exclusion: Kidney dysfunction (Creatinine Clearance <60 mL/min), liver enzymes >2x ULN, recent infection, or active smoking (confounds elastase levels).

Study Timeline (7 Days Total)

  • Days 1–3 (Baseline Phase): No drug. Establish the subject’s “normal” rate of elastin breakdown.
  • Days 4–6 (Intervention Phase): Active treatment.
  • Day 7 (Washout/Follow-up): Final measurements.

Dosing Regimen

  • Drug: Sivelestat Sodium Hydrate.
  • Dose: 300 mg dissolved in 100 mL saline.
  • Administration: IV infusion over 1 hour, administered twice daily (e.g., 8:00 AM and 8:00 PM).
    • Note: This approximates the total daily exposure of the standard ICU dose (0.2 mg/kg/hr $\times$70kg $\approx$ 336 mg/day) but concentrates it into pulses.

3. The Biomarkers (The “Readout”)

Since you cannot biopsy the lung or aorta, you must use a validated systemic surrogate.

Primary Endpoint: Urinary Desmosine/Creatinine Ratio (uDES/Cr)

  • Why: Desmosine and Isodesmosine are amino acids found only in cross-linked elastin. They are not reused by the body; when elastin breaks down, they are excreted in urine. They are the “gold standard” fluid biomarker for elastin degradation.
  • Method: LC-MS/MS (Liquid Chromatography-Tandem Mass Spectrometry). Do not use ELISA (lower specificity).
  • Sampling: First morning void is best, or 24-hour urine collection (more accurate but burdensome). For “simple,” use First Morning Void every day of the study.

Secondary Endpoints (Safety & Mechanism):

  • High-Sensitivity CRP (hs-CRP): To establish baseline systemic inflammation.
  • Neutrophil Elastase Activity: Measured in plasma (optional, difficult to stabilize ex-vivo).

4. Cost-Effective Execution Strategy

Conducting this in a medical school setting allows you to save costs on infrastructure while maintaining quality.

Item Strategy for Cost/Simplicity
Drug Sourcing Sivelestat is off-patent but not FDA-approved (available in Japan/China). A university pharmacy may need to import it as an investigational drug or compound it from research-grade API (requires strict purity testing/Certificate of Analysis).
Infusion Use the medical school’s Clinical Research Unit (CRU) or a day-stay unit. A nurse places a peripheral line (heplock) in the morning; the patient returns in the evening for the second dose (or keeps the line in for 3 days).
Labs Desmosine Testing: This is the most expensive assay. Send batched samples to a specialized core lab or a commercial partner like the Mayo Clinic Labs or specific research labs known for elastin research (e.g., labs studying COPD/Alpha-1 Antitrypsin).
Safety Basic Chem-7 and CBC before and after. Monitor blood pressure during infusion (transient hypotension is a rare side effect).

5. Scientific Reality Check (The Risk)

Before launching, you must consider the Signal-to-Noise Ratio problem.

  • The Problem: In a healthy person, Neutrophil Elastase activity is very low. Elastin has a half-life of 70+ years. The baseline desmosine excretion in a healthy male is low.
  • The Risk: Sivelestat works by inhibiting excess elastase (like in sepsis or acute lung injury). If the subject’s elastase is not elevated, the drug has no target to hit.
  • Result: You might see no change in desmosine, not because the drug failed, but because there was no pathological breakdown to inhibit.
  • Mitigation: Pre-screen the subject with a baseline uDES/Cr test. If it is at the bottom of the reference range, the trial is likely to yield a “null” result. If it is in the upper quartile (perhaps due to age/sub-clinical inflammation), the trial has a chance of success.

Protocol Summary for IRB/Committee

Title: Pilot Evaluation of Intermittent IV Sivelestat on Systemic Elastin Turnover in a Healthy Older Adult.

Design: N=1, Open-label, Pre-post intervention.

Intervention: Sivelestat 300mg IV q12h x 3 days.

Endpoint: Change in urinary Desmosine/Creatinine ratio (Day 1-3 mean vs. Day 4-6 mean).

Safety: Daily vitals, pre/post metabolic panel.

Based on the protocol designed in our previous step and the pricing data you provided, the direct acquisition cost for the medication is remarkably low.

For the N=1 study as suggested (300 mg per dose, twice daily, for 3 days), the estimated total medication cost is $630 – $702 USD.

Here is the breakdown of that calculation, along with a critical nuance regarding the dosing volume vs. cost.

1. The Calculation (Based on Proposed Protocol)

  • Vial Strength: 0.1 g (100 mg) per vial.
  • Dose Required: 300 mg per infusion.
  • Vials per Dose: 3 vials.
  • Frequency: Twice daily (Morning & Evening).
  • Daily Usage: 6 vials.
  • Duration: 3 Days.
  • Total Vials Needed: 18 vials.
Unit Cost (Approx) Total Calculation Total Cost (USD)
$35 / vial (Low est.) 18 vials $\times$ $35 $630
$39 / vial (High est.) 18 vials $\times$ $39 $702

seems like chondroitine supplement would be a good idea to antagonize ERC elastin receptor complex. Seems the only solution we have so far. Also, make a break for thoses as me who supplement with elastin or hyaluronic acid supplement ?..

1 Like

What is UBV?

Extra special typo. UVB. Fixed.

1 Like

Kamil is a longevity scientist in Bryan Kennedy’s lab at National University of Singapore, and brings up a point that we should consider (as we gobble down collagen):

Most ingested elastin or collagen is broken down to amino acids and very small peptides before absorption. A tiny fraction of elastin-derived dipeptides can enter the blood intact, but we currently have no evidence that the specific, pro-aging elastin fragments from the Nature Aging paper (e.g., VGVAPG “E-motif”) survive oral digestion and reach systemic levels comparable to those that shorten lifespan in mice. The risk signal is theoretical at this point, not demonstrated.

  • Yes, very small elastin-derived peptides (e.g., dipeptides) survive digestion and enter blood.
  • No current evidence that the pro-aging hexapeptide motifs like VGVAPG (E-motif) themselves are present in blood at significant levels after oral supplementation in humans; nobody has actually looked carefully.

See full analysis here: https://chatgpt.com/share/692bc5b8-f514-8008-a3ca-5cec35023acd