Category: 8. Health

  • Freezing molecules could halt the spread of brain cancer

    Freezing molecules could halt the spread of brain cancer

    Cancer research often reveals unexpected ways to fight disease by rethinking the cell’s environment. A new approach from the University of Cambridge suggests halting glioblastoma, the deadliest brain cancer, by freezing the very molecules that enable its spread.

    Instead of killing cells outright, this strategy changes the conditions around them, turning aggressive invaders into quiet neighbors.


    Glioblastoma is the most common brain cancer and one of the hardest to treat. Patients often survive less than 15 months after diagnosis, with only 15 percent living beyond five years. Surgery and radiotherapy can slow progress but rarely prevent recurrence.

    Cancer cells left behind infiltrate healthy brain tissue, spreading silently through microscopic channels. Current drugs also struggle to penetrate the dense tumor mass, leaving few effective options.

    Cancer uses hyaluronic acid

    At the heart of this discovery is hyaluronic acid (HA), a sugar-like polymer that shapes much of the brain’s supporting structure.

    Cancer cells rely on HA’s flexibility to latch onto surface receptors, especially CD44, which drives their movement. By chemically locking HA in place, scientists stripped away its flexibility. This effectively reprogrammed glioblastoma cells into dormancy, preventing them from invading new tissue.

    Study co-author Melinda Duer is a professor in the Yusuf Hamied Department of Chemistry at the University of Cambridge.

    “Fundamentally, hyaluronic acid molecules need to be flexible to bind to cancer cell receptors,” said Professor Duer.

    “If you can stop hyaluronic acid being flexible, you can stop cancer cells from spreading. The remarkable thing is that we didn’t have to kill the cells – we simply changed their environment, and they gave up trying to escape and invade neighbouring tissue.”

    Freezing flexibility stops cancer

    The Cambridge study confirmed that HA’s behavior is not just about molecular size but about motion. High-molecular-weight HA, usually linked with healthy tissue, can still promote cancer invasion when diluted.

    At lower concentrations, HA molecules move freely, adopting shapes that fit tightly into CD44 binding sites. This triggers powerful invasion signals.

    At higher concentrations, HA molecules entangle with each other, limiting flexibility and dampening those signals.

    Molecular motion and signals

    Nuclear magnetic resonance spectroscopy revealed that HA’s ability to twist into specific conformations within nanoseconds determines whether it binds strongly to CD44.

    When flexible, HA activates pathways that push cells to migrate. When immobilized, HA blocks these signals.

    This explains why swelling in the brain after surgery, which dilutes HA, can encourage cancer to return at the surgical site.

    Protein responses to hyaluronic acid

    Proteomic analysis of glioblastoma cells highlighted how HA concentration reshapes their internal machinery.

    In flexible environments, cells developed star-shaped structures and invasive protrusions, supported by actin-bundling proteins and enzymes that degrade tissue.

    In stiffer environments, those invasive proteins disappeared. Instead, cells activated survival pathways, entered a dormant state, and upregulated proteins linked to quiescence, including Notch-2.

    This switch showed how extracellular conditions can push cancer cells either into attack mode or into stillness.

    Freezing molecules stops cancer

    Researchers tested this idea further by creating a chemically modified HA that crosslinks tightly into the extracellular matrix. This version lost flexibility and, when added to cell cultures, completely stopped invasion.

    Cells responded in the same way as in dense HA conditions, showing altered protein expression and increased dormancy signals. This provided strong evidence that flexibility itself, not size, is the decisive factor in driving cancer spread.

    The findings give a new explanation for glioblastoma’s notorious tendency to regrow after surgery. Fluid build-up, or oedema, dilutes HA at the surgical site, making it more flexible and more capable of binding CD44.

    This diluted environment inadvertently encourages cancer cells to invade again. By freezing HA in place, scientists hope to counteract this effect and stop recurrence before it starts.

    Freezing may help treat cancer

    “This could be a real opportunity to slow glioblastoma progression,” said Duer. “And because our approach doesn’t require drugs to enter every single cancer cell, it could in principle work for many solid tumours where the surrounding matrix drives invasion.

    “Cancer cells behave the way they do in part because of their environment. If you change their environment, you can change the cells.”

    The work now moves into animal testing before any patient trials. Still, the implications extend beyond brain cancer. Many tumors rely on signals from their surrounding environment to spread.

    By targeting flexibility in key molecules like HA, researchers may uncover a new class of treatments that turn cancer’s supportive scaffolding into a trap. Instead of chasing runaway cells, this method locks the very ground beneath them.

    The study is published in the journal Royal Society Open Science.

    —–

    Like what you read? Subscribe to our newsletter for engaging articles, exclusive content, and the latest updates. 

    Check us out on EarthSnap, a free app brought to you by Eric Ralls and Earth.com.

    —–

    Continue Reading

  • This Type of Plant-Based Diet Raises Heart Disease Risk

    This Type of Plant-Based Diet Raises Heart Disease Risk

    • New research suggests not all plant-based diets are healthy.
    • Some plant-based diets may be bad for heart health.
    • Experts suggest focusing on certain elements when eating a plant-based diet.

    Many medical organizations recommend following a plant-forward diet, but new research points out that all plant-based foods are not created equal. In fact, some may even be bad for your heart health.

    The study, which was published in the American Journal of Preventive Cardiology, found a clear link between eating an unhealthy plant-based diet and a key biomarker linked to having a heart attack. For the study, researchers analyzed data from more than 7,700 people who participated in the 1999-2004 National Health and Nutrition Examination Survey (NHANES). (Yes, that’s data from more than 20 years ago—but experts say the findings still apply.) All of the participants were at least 20 years old and had no history of heart disease at the start of the study.

    During the study period, the participants reported what they ate over a 24-hour period and gave blood samples to allow the researchers to look for biomarkers of heart disease. The researchers discovered that people who followed a healthy plant-based diet had a 49% lower risk of having higher levels of cardiac troponin I. (Cardiac troponin I is a sign of cardiac muscle damage and is linked to heart attack risk.)

    Meet the experts: Scott Keatley, R.D., is co-owner of Keatley Medical Nutrition Therapy; Hosam Hmoud, M.D., cardiology fellow at Northwell’s Lenox Hill Hospital; Dena Champion, R.D., a dietitian at The Ohio State University Wexner Medical Center; Jennifer Wong, M.D., a cardiologist and medical director of Non-Invasive Cardiology at MemorialCare Heart and Vascular Institute at Orange Coast Medical Center in Fountain Valley, CA.

    But people who followed an unhealthy plant-based diet had a 65% higher risk of having elevated levels of cardiac troponin I, meaning they had a greater heart attack risk. “Supporting access to and adoption of healthy plant-based diets may be a useful strategy for promoting population-level cardiovascular health,” the researchers wrote in the conclusion.

    The study raises a lot of questions about what qualifies as a “healthy” or “unhealthy” plant-based diet, plus the potential impact on your heart. Here’s what cardiologists and dietitians want you to know.

    Why are certain plant-based foods bad for your heart?

    It’s easy to confuse “plant-based” with “healthy,” but they’re not the same thing, points out Dena Champion, R.D., a dietitian at The Ohio State University Wexner Medical Center. “The term ‘plant-based’ lacks a universal definition, which creates confusion for consumers,” she says. “Many food manufacturers and restaurants use ‘plant-based’ simply to indicate their products contain no animal ingredients.”

    Having a lack of animal products doesn’t automatically make a food healthy, Champion points out. Scott Keatley, R.D., co-owner of Keatley Medical Nutrition Therapy, agrees. “Not all plant-based foods are created equal,” he says. “A diet heavy in refined grains, fried foods, sugary beverages, and ultra-processed snacks may technically be ‘plant-based,’ but it lacks the protective compounds that whole fruits, vegetables, legumes, and intact grains provide.”

    These unhealthy plant foods can spike your blood sugar, contribute to bodily inflammation, contain trans fats, and deliver excess calories without important nutrients, raising the risk of heart disease and other health issues, Keatley says.

    “It’s important to point out that a vegan or vegetarian diet can absolutely be a heart-healthy diet, but eliminating animal products alone does not make a diet healthy,” Champion says.

    How to tell when plant-based foods are unhealthy

    There are a few different ways to tell if a plant-based food is unhealthy, according to Keatley: the level of processing and the amount of sodium and additives.

    “The further a food is from its natural state, the more likely it has lost beneficial nutrients while gaining added sugars, unhealthy fats, or refined starches,” Keatley explains. “Packaged plant-based meals, snacks, or meat alternatives often carry sodium levels comparable to processed meats and may include stabilizers and oils that don’t support cardiovascular health.”

    Champion also recommends being on the lookout for higher amounts of saturated fat, and making sure to read the nutrition label carefully. “The label ‘plant-based’ does not necessarily indicate anything about the healthiness of a food,” she stresses.

    How to follow a heart-healthy diet

    In general, whole foods—not packaged products—are best for your heart health, according to Jennifer Wong, M.D., a cardiologist and medical director of Non-Invasive Cardiology at MemorialCare Heart and Vascular Institute at Orange Coast Medical Center in Fountain Valley, CA. “I recommend the DASH Diet,” she says. “It’s low in salt, high in fruits and vegetables, whole grains, and legumes. It focuses on healthier fats like olive oil and avocado rather than animal fats.”

    But the Mediterranean diet is also helpful for heart health, says Hosam Hmoud, M.D., cardiology fellow at Northwell’s Lenox Hill Hospital. “I encourage all of my patients to follow a Mediterranean-style diet which consists of whole grains, legumes, fruits, vegetables, and lean meats,” he says.

    Ultimately, Keatley suggests filling your plate with whole foods first and building out your diet from there. “Most people already consume a plant-based diet if you look closely—grains, fruits, vegetables, and even snack foods all come from plants,” he points out. “The real distinction isn’t whether something is plant-based or not, but whether it’s minimally processed and nutrient-dense.”

    Continue Reading

  • The aging factor EPS8 induces disease-related protein aggregation through RAC signaling hyperactivation

    The aging factor EPS8 induces disease-related protein aggregation through RAC signaling hyperactivation

    C. elegans strains

    C. elegans strains were cultured at 20 °C on standard Nematode Growth Medium seeded with OP50 Escherichia coli70. On day 1 of adulthood, worms were transferred to plates containing OP50 E. coli (or HT115 E. coli for RNAi experiments) supplemented with 100 μg ml−1 5-fluoro-2′-deoxyuridine to prevent progeny development, except in lifespan assays. All experiments were conducted using hermaphrodite worms, and the age of the worms is indicated in the corresponding figures and figure legends.

    WT (N2) and AM141 (rmIs133[unc-54p::Q40::YFP]) strains were obtained from the Caenorhabditis Genetics Center (CGC), supported by the National Institutes of Health Office of Research Infrastructure Programs (P40 OD010440). RB751 (eps-8(ok539)) was generated by the C. elegans Gene Knockout Consortium and acquired from the CGC. AM23 (rmIs298[F25B3.3p::Q19::CFP]) and AM716 (rmIs284[F25B3.3p::Q67::YFP]) strains were gifted by Richard I. Morimoto24. MAH602 (sqIs61[vha-6p::Q44::YFP + rol-6(su1006)]) was provided by Malene Hansen71. ZM5838 (hpIs223[rgef-1p::FUSWT::GFP]), ZM5844 (hpIs233[rgef-1p::FUSP525L::GFP]) and ZM5842 (hpIs228[rgef-1p::FUSR522G::GFP]) were provided by Peter St. George-Hyslop45. CK405(Psnb-1::TDP-43WT,myo-2p::dsRED) and CK423 (Psnb-1::TDP-43M337V,myo-2p::dsRED) were provided by Brian C. Kraemer33.

    From these strains, we generated NFB2862 (Psnb-1::TDP-43WT,myo-2p::dsRED;juIs76[unc-25p::GFP + lin-15(+)]II) and NFB2863 (Psnb-1::TDP-43M337V,myo-2p::dsRED;juIs76[unc-25p::GFP + lin-15(+)]II). NFB2858 (rmIs298[F25B3.3p::Q19::CFP];otIs549[unc-25p::unc-25(partial)::mChopti::unc-54 3′ untranslated region (UTR) + pha-1(+)];him-5(e1490)V), NFB2859 (rmIs284[F25B3.3p::Q67::YFP];otIs549[unc-25p::unc-25(partial)::mChopti::unc-54 3′ UTR + pha-1(+)];him-5(e1490)V), NFB2860 (hpIs223[rgef-1p::FUSWT::GFP];otIs549[unc-25p::unc-25(partial)::mChopti::unc-54 3′ UTR + pha-1(+)];him-5(e1490)V) and NFB2861 (hpIs233[rgef-1p::FUSP525L::GFP];otIs549[unc-25p::unc-25(partial)::mChopti::unc-54 3′ UTR + pha-1(+)];him-5(e1490)V) were generated by crossing the respective polyQ and FUS-expressing strains with the OH13526 strain72. For RNAi in the neurons of polyQ67 worms, we used the DVG196 strain (rmIs284[F25B3.3p::Q67::YFP];sid-1(pk3321)V;uIs69[pCFJ90(myo-2p::mCherry) + unc-119p::sid-1]).

    Worms expressing endogenous WT EPS-8::3xHA (VDL05, eps-8(syb2901)IV) or mutant EPS-8(K524R/K583R/K621R::3×HA) (VDL06, eps-8(syb2901, syb3149)IV) were previously generated via CRISPR–Cas9 (ref. 14). The strains DVG344 (rmIs284[pF25B3.3::Q67::YFP]);eps-8(syb2901) and DVG363 (rmIs133[unc-54p::Q40::YFP]);eps-8(syb2901) were generated by crossing VDL05 with AM716 and AM141, respectively. DVG345 (rmIs284[pF25B3.3::Q67::YFP]);eps-8(syb2901, syb3149) and DVG364 (rmIs133[unc-54p::Q40::YFP]);eps-8(syb2901, syb3149) were generated by crossing VDL06 to AM716 and AM141, respectively. These strains were validated by sequencing using the following primers: eps-8(syb2901): 5′-TTTGTTCGAAGCATGAACGA-3′ and 5′-AGCAGCCCCTGAAATAGTGA-3′; eps-8(syb2901, syb3149): 5′-AACGAGCTAGCAATCCGAAA-3′ and 5′-AGTGCTCTGCCGTCATTAAT-3′. DVG365 (rmIs284[pF25B3.3::Q67::YFP];eps-8(ok539)) was generated by crossing RB751 to AM716. The strain was outcrossed two times to AM716 and validated by polymerase chain reaction with 5′-TCTCCACCACCACAACGTAA-3′ and 5′-GCGGAGCAACTCTTCCATAG-3′ primers.

    RNAi constructs

    Adult worms were fed HT115 E. coli carrying either an empty control vector (L4440) or vectors expressing double-stranded RNAi. The RNAi constructs targeting eps-8, ifb-2, jnk-1, kgb-1, mig-2 and otub-3 were obtained from the Vidal library. The csn-6, F07A11.4, math-33, rac-2, usp-4, usp-5 and usp-48 RNAi constructs were obtained from the Ahringer library. All RNAi constructs were sequence verified. The RNAi sequences are listed in Supplementary Table 2.

    Lifespan assay

    Larvae were synchronized using the egg-laying protocol and grown on OP50 E. coli at 20 °C until day 1 of adulthood. Adult hermaphrodites were then transferred to plates with HT115 E. coli carrying either an empty vector or RNAi constructs for lifespan assays. All lifespan assays were performed at 20 °C. Each condition included 96 worms, scored daily or every other day73. Worms that were lost, burrowed into the medium, had a protruding vulva or underwent bagging were censored73.

    Nose touch assay

    Age-synchronized worms were assessed for nose touch response as previously described74,75,76. In brief, worms were placed on a thin bacterial lawn, and an eyelash pick was positioned in front of a forward-moving animal. A lack of response was recorded when the worm continued moving forward to crawl under or over the pick. For each condition, 30–40 animals were tested by monitoring the number of responses to a total of 10 gentle eyelash touches.

    Chemotaxis assay

    Freshly prepared agar plates (2% agar, 5 mM KPO4 (pH 6.0), 1 mM CaCl2, 1 mM MgSO4) were divided into four equal quadrants, along with an inner circle measuring approximately 1 cm across diagonally. A test solution (0.5% benzaldehyde (Sigma-Aldrich, B1334) in ethanol + 0.25 M sodium azide) and a control solution (ethanol + 0.25 M sodium azide) were added to two opposing diagonal quadrants. On the indicated days of adulthood (as shown in the corresponding figures), worms were collected in S-Basal medium, washed three times to remove residual bacteria and placed at the center of the chemotaxis plate. The plates were sealed with parafilm and incubated at 20 °C for 90 minutes. The number of worms in each quadrant was counted, excluding those that did not cross the inner circle. The chemotaxis index was calculated using the following formula: chemotaxis index = ((number of animals in test quadrants) − (number of animals in control quadrants)) / total number of animals77.

    Motility assays

    C. elegans were synchronized on OP50 E. coli using the egg-laying method and grown until day 1 of adulthood and then randomly transferred to plates with HT115 E. coli containing either empty vector or RNAi for the remainder of the experiment. For experiments with Ub-less EPS-8 mutants or DUB inhibitor treatment, worms were instead transferred to fresh plates containing OP50 E. coli. On the indicated day of adulthood (as shown in the corresponding figures), worms were randomly picked and transferred to a drop of M9 buffer, allowing 30 seconds for recovery24. Body bends were then recorded for 30 seconds and analyzed using ImageJ software (version 1.53k) with the wrMTrck plugin (https://www.phage.dk/plugins/)78,79. The locomotion velocity data were used to calculate body bends per second.

    Microscopy

    For imaging GABAergic neurons, fluorescent reporter worms were anesthetized with a drop of 0.5 M sodium azide (Sigma-Aldrich, 26628-22-8) on 4% agarose pads (diluted in distilled water) placed over a standard microscope glass slide (Rogo-Sampaic, 11854782). These preparations were sealed with 24 × 60-mm coverslips (RS France, BPD025). To score the number of GABAergic neurons and ventral nerve cord projections, we used a Zeiss Axio Imager.M2 microscope with a ×40 objective. Whole-body worm images were acquired using a Leica THUNDER Imager microscope with Tile Scan function and a ×40 objective.

    Human cell lines

    HEK293T/17 cells (American Type Culture Collection (ATCC), CRL-11268) were plated on 0.1% gelatin-coated plates and grown in DMEM (Thermo Fisher Scientific, 11966025), supplemented with 1% MEM non-essential amino acids (Thermo Fisher Scientific, 11140035), 1% GlutaMAX (Life Technologies, 35050038) and 10% FBS (Thermo Fisher Scientific, 10500064) at 37 °C with 5% CO2. ALS-iPSCs (FUSP525L/P525L) were kindly provided by Irene Bozzoni and Alessandro Rosa37. iPSCs were cultured on Geltrex (Thermo Fisher Scientific, A1413302) using mTeSR1 medium (STEMCELL Technologies, 85850) at 37 °C with 5% CO2. All cell lines were routinely tested for mycoplasma contamination, and no contamination was detected.

    Motor neuron differentiation

    Motor neurons were derived from ALS-iPSCs using a monolayer-based differentiation protocol80. ALS-iPSCs were seeded on Geltrex-coated plates and maintained in mTeSR1 medium until confluent. Differentiation was initiated using neuron differentiation medium composed of DMEM/F12 and Neurobasal (1:1; Thermo Fisher Scientific, 11330057 and 21103049), supplemented with non-essential amino acids, GlutaMAX (Thermo Fisher Scientific, 35050038), B27 (Thermo Fisher Scientific, 12587010) and N2 (Thermo Fisher Scientific, 17502048).

    From day 0 to day 6, the medium was further supplemented with 1 μM retinoic acid (Sigma-Aldrich, R2625), 1 μM smoothened agonist (SAG; Sigma-Aldrich, 566661), 0.1 μM LDN-193189 (Miltenyi Biotec, 130-103-925) and 10 μM SB-431542 (Miltenyi Biotec, 130-105-336). From day 7 to day 14, the neuron differentiation media were supplemented with 1 μM retinoic acid, 1 μM SAG, 4 μM SU-5402 (Sigma-Aldrich, SML0443) and 5 μM DAPT (Sigma-Aldrich, D5942). After day 14, differentiated motor neurons were dissociated and replated on poly-l-ornithine (Sigma-Aldrich, P3655) and laminin-coated (Thermo Fisher Scientific, 23017015) plates in Neurobasal medium, supplemented with non-essential amino acids, GlutaMAX, N2, B27 and neurotrophic factors (10 ng ml−1 BDNF (BIOZOL, 450-02) and 10 ng ml−1 GDNF (BIOZOL, 450-10)).

    Lentiviral infection of human cells

    Lentivirus (LV)-non-targeting short hairpin RNA (shRNA), LV-EPS8 shRNA 1 (TRCN0000061544), LV-EPS8 shRNA 2 (TRCN0000061545), LV-USP4 shRNA 1 (TRCN0000004039) and LV-USP4 shRNA 2 (TRCN0000004040) in the pLKO.1-puro backbone were obtained from Mission shRNA (Sigma-Aldrich). Supplementary Table 2 contains the target sequences of each shRNA construct.

    To generate stable shRNA-expressing HEK293 cell lines, cells were transduced with 5 µl of concentrated lentivirus and selected with 2 µg ml−1 puromycin (Thermo Fisher Scientific, A1113803). For lentiviral infection of iPSCs, cells were dissociated using Accutase (Thermo Fisher Scientific, A1110501), and 100,000 cells were seeded on Geltrex-coated plates in mTeSR1 medium supplemented with 10 μM ROCK inhibitor for 1 day. The next day, cells were infected with 5 µl of concentrated lentivirus. Medium was replaced the following day to remove residual virus. Selection for lentiviral integration was performed using 2 µg ml−1 puromycin for 2 days.

    Transfection of HEK293 cells

    HEK293 cells (ATCC, CRL-11268) were seeded on 0.1% gelatin-coated plates. When cells reached approximately 40% confluency, they were transfected with 1 μg of one of the following plasmids using FuGENE HD (Promega), according to the manufacturer’s instructions: pARIS-mCherry-httQ23-GFP, pARIS-mCherry-httQ100-GFP, pLVX-Puro-TDP-43-WT, pLVX-Puro-TDP-43-A382T, pcDNA3.1-FUS-HA-WT or pcDNA3.1-FUS-HA-P525L. In the indicated experiments, cells were co-transfected with an additional 1 μg of the pCMV3-EPS8-HA plasmid. The cells were collected after 72 hours of incubation in standard medium. The pARIS-mCherry-httQ23-GFP and pARIS-mCherry-httQ100-GFP plasmids were generously provided by Frédéric Saudou81. The FUS-HA-WT and FUS-HA-P525L plasmids were a gift from Dorothee Dormann82. The pLVX-Puro-TDP-43-WT and pLVX-Puro-TDP-43-A382T plasmids were provided by Shawn Ferguson (Addgene, 133753 and 133756)83. The pCMV3-EPS8-HA plasmid was obtained from Sino Biological (HG11153-CY).

    Filter trap and western blot

    For filter trap assays, synchronized adult C. elegans were collected and washed with M9 buffer, and worm pellets were snap frozen in liquid nitrogen. Frozen pellets were thawed on ice and lysed in non-denaturing buffer (50 mM HEPES (pH 7.4), 150 mM NaCl, 1 mM EDTA, 1% Triton X-100, 2 mM sodium orthovanadate, 1 mM PMSF, protease inhibitor cocktail (Roche)) using a Precellys 24 homogenizer. Lysates were cleared of worm debris by centrifugation (8,000g, 5 minutes, 4 °C), and protein concentrations were determined using the BCA assay (Thermo Fisher Scientific). To assess protein levels by western blot, 30 μg of total protein was separated by SDS-PAGE and transferred to polyvinylidene difluoride membranes (Millipore). To assess aggregated proteins by filter trap, 100 μg of total protein was supplemented with SDS to a final concentration of 0.5% and loaded onto a cellulose acetate membrane assembled in a slot-blot apparatus (Bio-Rad). Then, the membrane was washed with 0.2% SDS, and SDS-resistant aggregates were detected by immunoblotting.

    If lysates were used solely for western blot, worms were lysed with a Precellys 24 homogenizer in buffer containing 50 mM Tris-HCl (pH 7.8), 150 mM NaCl, 1% Triton X-100, 0.25% sodium deoxycholate, 1 mM EDTA, 25 mM N-ethylmaleimide, 2 mM sodium orthovanadate, 1 mM PMSF and protease inhibitor cocktail. Lysates were cleared at 10,600g for 10 minutes at 4 °C, and 30 μg of protein was used for western blot experiments. For analysis of polyQ monomers and SDS-insoluble polyQ aggregates, age-synchronized worms were lysed by sonication in native buffer (50 mM Tris (pH 8), 150 mM NaCl, 5 mM EDTA, 1 mM PMSF, protease inhibitor cocktail). Then, 30 μg of total protein was mixed with SDS to a final concentration of 0.4% and resolved by 12.5% SDS-PAGE.

    For both filter trap and western blot analyses of C. elegans, immunoblotting was performed with antibodies against GFP (AMSBIO, TP401, dilution 1:5,000), FUS (Abcam, ab154141, clone CL0190, 1:1,000) and TDP-43 (Abcam, ab225710, 1:1,000). Additionally, for western blot experiments, immunoblotting was conducted with anti-EPS8L2 (Abcam, ab85960, 1:1,000), anti-LGG-1 (ref. 84, 1:2,000) and α-tubulin (Sigma-Aldrich, T6199, 1:5,000).

    For filter trap and western blot analysis of HEK293 cell lines, the cells were collected in lysis buffer (50 mM HEPES (pH 7.4), 150 mM NaCl, 1 mM EDTA, 1% Triton X-100, 2 mM sodium orthovanadate, 1 mM PMSF, protease inhibitor cocktail), followed by homogenization through a 27-gauge syringe needle. Lysates from cells expressing pARIS-mCherry-httQ23-GFP, pARIS-mCherry-httQ100-GFP or without any overexpression were centrifuged at 8,000g for 5 minutes at 4 °C. Lysates from cells expressing FUS-HA-WT, FUS-HA-P525L, pLVX-Puro-TDP-43-WT or pLVX-Puro-TDP-43-A382T were centrifuged at 1,000g for 5 minutes at 4 °C. The supernatants were collected, and protein concentrations were measured with the BCA assay. For western blot, 30 μg of protein was analyzed as above. For filter trap analysis, 100 μg of total protein was supplemented with SDS to a final concentration of 0.5% and loaded onto a cellulose acetate membrane assembled in a slot-blot apparatus as described above. The membrane was then washed with 0.2% SDS, and SDS-resistant protein aggregates were evaluated by immunoblotting. For filter trap analysis, immunoblotting was conducted with antibodies against GFP (AMSBIO, TP401, 1:5,000), FUS (Abcam, ab154141, clone CL0190, 1:1,000) and TDP-43 (Abcam, ab225710, 1:1,000). For western blot, immunoblotting was conducted with anti-EPS8 (Proteintech, 12455-1-AP, 1:1,000), anti-β-actin (Abcam, 8226, 1:5,000), anti-HTT (Cell Signaling Technology, 5656, 1:1,000), FUS (Abcam, ab154141, clone CL0190, 1:1,000), TDP-43 (Abcam, ab225710, 1:1,000), anti-LC3B (Cell Signaling Technology, 2775, 1:1,000) and anti-USP-4 (Abcam, ab181105, 1:1,000).

    For necroptosis analysis, iPSC-derived motor neurons were lysed in RIPA buffer (50 mM Tris-HCl (pH 7.4), 150 mM NaCl, 1% Triton X-100, 1% sodium deoxycholate, 0.1% SDS, 1 mM EDTA, 1 mM PMSF, protease inhibitor cocktail). Immunoblotting was performed using anti-phospho-RIP (Ser166) (Cell Signaling Technology, 65746, clone D1L3S, 1:1,000) and anti-RIP (Cell Signaling Technology, 3493, clone D94C12, 1:1,000). Densitometry of filter trap and western blot assays was performed using ImageJ software (version 1.51).

    Protein immunoprecipitation for interaction analysis

    HEK293 cells were collected and lysed in a protein lysis buffer containing 50 mM Tris-HCl (pH 6.7) 150 mM NaCl, 1% NP40, 0.25% sodium deoxycholate, 1 mM EDTA, 1 mM PMSF, 1 mM sodium orthovanadate, 1 mM NaF and protease inhibitor cocktail. Lysates were homogenized through a 27-gauge syringe needle and centrifuged at 13,000g for 15 minutes at 4 °C. Supernatants were incubated on ice for 1 hour with anti-USP-4 antibody (Abcam, ab181105, 1:100). As a negative control, the same amount of protein was incubated with anti-normal rabbit IgG (Cell Signaling Technology, 2729, 1:378). Samples were then incubated with 50 µl of µMACS MicroBeads for 1 hour at 4 °C with overhead shaking. Then, the samples were loaded onto pre-cleared µMACS columns (Miltenyi Biotec, 130-042-701). The beads were washed three times with a buffer containing 50 mM Tris (pH 7.5), 150 mM NaCl, 5% glycerol and 0.05% Triton, followed by five washes with 50 mM Tris (pH 7.5) and 150 mM NaCl. The samples were eluted with 75 μl of boiled 2× Laemmli buffer, boiled for 5 minutes at 95 °C and analyzed by western blotting.

    Native gels analysis

    HEK293 cells expressing CMV:mRFP-Q74 (ref. 30) were lysed in buffer containing 50 mM Tris-HCl (pH 7.4), 150 mM NaCl, 0.5% NP-40, 2 mM EDTA, 1 mM EGTA, 10% glycerol, 2 mM sodium orthovanadate, 1 mM PMSF and protease inhibitor cocktail. Lysates were homogenized using a 27-gauge syringe needle and centrifuged at 12,000g for 15 minutes at 4 °C. Supernatants were collected, and protein concentrations were determined using the BCA protein assay (Thermo Fisher Scientific). Equal amounts of protein lysates were mixed 1:1 with sample buffer (50 mM Tris-HCl (pH 6.8), 10% glycerol, 0.01% bromophenol blue). Then, 20 μg of total protein was separated using 4–15% Tris-Glycine eXtended protein gels (Bio-Rad) and imaged via fluorescence using LICOR Odyssey M.

    Immunocytochemistry

    Cells were fixed with 4% paraformaldehyde in PBS for 20 minutes, followed by permeabilization with 0.2% Triton X-100 in PBS (10 minutes) and blocking with 3% BSA in 0.2% Triton X-100 in PBS (10 minutes). The cells were then incubated with anti-MAP2 (Sigma-Aldrich, M1406, 1:300) and rabbit anti-cleaved caspase-3 (Cell Signalling Technology, 9661S, 1:300) for 2 hours at room temperature. After washing with PBS, cells were incubated with secondary antibodies (Alexa Fluor 488 goat anti-mouse (Thermo Fisher Scientific, A-11029, 1:500) and Alexa Fluor 568 F(ab′)2 fragment of goat anti-rabbit IgG (H + L) (Thermo Fisher Scientific, A-21069, 1:500)) and Hoechst 33342 (Life Technologies, 1656104) for 1 hour at room temperature. Finally, the coverslips were rinsed in PBS, followed by a distilled water wash, and then mounted onto microscope slides with FluorSave Reagent (Merck, 345789).

    CytoD, RAC activator and DUB inhibitor treatment

    For CytoD treatment, worms were collected and randomly divided equally into M9 solutions containing either 10 μM CytoD (STEMCELL Technologies, 100-0557) or DMSO as a vehicle control. The worms were incubated with CytoD or DMSO for 6 hours on a shaker. For DUB inhibitor experiments, worms were collected and randomly transferred onto plates with OP50 bacteria covered with a final concentration of 13.7 μg ml−1 PR-619 (Merck, 662141) or vehicle control (DMSO) for either 4 hours or 1 day as indicated in the corresponding figures.

    HEK293 cells were treated with 2 μM CytoD or DMSO for 4 hours before lysis. For RAC activation, cells were treated with 2 U ml−1 Rac/Cdc42 Activator II (Cytoskeleton, CN02-A) for 6 hours.

    Proteasome activity

    Day 5 adult worms and HEK293 cells were lysed in proteasome activity assay buffer (50 mM Tris-HCl (pH 7.5), 10% glycerol, 5 mM MgCl2, 0.5 mM EDTA, 2 mM ATP, 1 mM DTT) using a Precellys 24 or a 27-gauge syringe, respectively. The samples were centrifuged at 10,000g for 10 minutes at 4 °C, and the supernatants were collected. Protein concentrations were determined using the BCA protein assay kit.

    To measure chymotrypsin-like proteasome activity, 25 μg of total protein was incubated with the fluorogenic substrate Suc-Leu-Leu-Val-Tyr-AMC (Enzo Life Sciences, BML-P802) in 96-well plates (BD Falcon). Fluorescence was measured every 5 minutes for 2 hours at 20 °C (C. elegans) or 37 °C (human cells) using a microplate fluorometer (PerkinElmer, EnSpire) at 380-nm excitation and 460-nm emission.

    Statistics and reproducibility

    For quantification of filter trap and western blot data, we presented the results as relative changes compared with the corresponding control conditions. To average and statistically analyze independent experiments for these assays, we normalized test conditions to their corresponding control groups measured concurrently in each replicate experiment. Given that all the control groups were set to 100, we used a non-parametric Wilcoxon test when comparing two conditions to assess changes in protein aggregation and protein levels. For all other assays, we used parametric tests. Data distribution was assumed to be normal, but this was not formally tested. When more than two conditions or two independent variables were compared, we used one-way or two-way ANOVA followed by multiple comparisons tests. All statistical analyses were performed using GraphPad Prism (version 10.4.1).

    For lifespan experiments, we used GraphPad Prism (version 10.4.1) and OASIS (version 1)85 to determine median and mean lifespan, respectively. The P values were calculated using the log-rank (Mantel–Cox) method and refer to comparisons between experimental and control animals within a single lifespan experiment. Each lifespan graph represents a single, representative experiment. Supplementary Table 1 contains the number of total/censored worms as well as detailed statistical analyses for each replicate lifespan experiment.

    No statistical methods were used to predetermine sample size, but our sample sizes are similar to, or greater than, those reported in previous publications using the same procedures9,14,16,26,30,33,44,46,50,73,75,76,78,86,87,88. For motility assays, worms were excluded from analysis if they showed fewer than 0.1 body bends per second or were not recognized by the ImageJ software. No animals or data points were excluded from other analyses. For lifespan assays, worms were randomly picked and transferred from the synchronized population to the different experimental conditions. For all other experiments, worms were randomly distributed into the various experimental groups from single pulls of synchronized populations. Human cells were distributed to the various groups of all experiments from single pulls. Data collection was not randomized. Data collection and analysis were not performed blinded to the conditions of the experiments.

    Reporting summary

    Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.

    Continue Reading

  • Management of Open Tibial Fractures: Potential Treatment Recommendatio

    Management of Open Tibial Fractures: Potential Treatment Recommendatio

    1Medical School, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa; 2Department of Orthopaedic Surgery and Sports Medicine, Burjeel Hospital for Advanced Surgery, Dubai, United Arab Emirates; 3Division of Orthopaedic Surgery, Groote Schuur Hospital, Cape Town, South Africa; 4Orthopaedic Research Unit, University of Cape Town, Cape Town, South Africa; 5Department of Orthopaedic Surgery, The Royal Brisbane and Women’s Hospital, Brisbane, Australia; 6Faculty of Medicine, Biruni University, Istanbul, Turkey

    Correspondence: Erik Hohmann, Burjeel Hospital for Advanced Surgery, Dubai, United Arab Emirates, Email [email protected]

    Abstract: Treatment guidelines for open tibial fractures are well established in high-income countries, but their implementation in low-resource settings remains challenging. To date, only one African country has attempted to formulate national, consensus-based guidelines that cover key aspects such as antibiotic administration, initial stabilization, surgical debridement, wound management, and definitive fixation. This narrative review summarizes current principles for the initial management of open tibial shaft fractures and evaluates their relevance in the Southern African context, particularly in rural and resource-constrained environments. Given the limited availability of advanced imaging, specialized implants, and soft-tissue coverage expertise, clinicians must adapt existing protocols to local capacities. This review highlights areas where treatment recommendations from high-income settings may not be feasible and identifies practical considerations for applying initial management principles in Southern Africa. The need for context-specific, resource-appropriate guidelines is emphasized.

    Keywords: open tibial fractures, initial management, low-resource countries, Southern Africa, review

    Introduction

    The treatment of open tibial shaft fractures presents a significant challenge due to limited soft tissue coverage and compromised blood supply.1 Due to its close proximity to the skin, the tibia is particularly vulnerable to becoming an open fracture with significant soft tissue damage, often resulting in complications such as infection and non-union.2 The primary goals of treatment are to promote an optimal environment for fracture healing, minimize complications, and restore limb function as effectively as possible.3 Achieving these goals can pose significant challenges for both patients and healthcare systems.3

    The healthcare system in Southern Africa consists of both public and private sectors, with marked inequalities in access and quality of care.4,5 Public healthcare, largely financed by government funding, serves the majority (particularly those in rural and economically disadvantaged areas) but often suffers from resource limitations, including staff shortages, inadequate infrastructure, and irregular medication supply.4,5 In contrast, private healthcare is supported by insurance or out-of-pocket payments and delivers superior services, though it is primarily accessible to wealthier, urban populations.4,5 In South Africa, for example, a dual healthcare model exists where approximately 80% of the population depends on the public sector, while the majority of resources are concentrated in the private sector, which caters to just 20% of citizens.4,5 Therefore, the management of open fractures in low to middle-income countries of Africa presents unique challenges, including limited early access to specialist care, delays in the administration of intravenous antibiotics, difficulties with choosing appropriate methods of fixation and wound closure, as well as patients’ health-seeking behaviours prior to accessing formal orthopaedic care.6 In addition, access to healthcare remains a significant issue, particularly in rural areas outside urban centres.7

    Open fractures should be managed using a standardized care pathway that includes the prompt administration of antibiotics, surgical debridement to remove all contaminated and devitalized tissue, thorough irrigation of the wound in the operating theatre, and fracture stabilization using either internal fixation, such as intramedullary nailing, or external fixation.8–11 While these guidelines are universally applicable, their implementation may not be practical or feasible in low-income countries. The Malawi Orthopaedic Association/AO Alliance has published a national consensus statement, outlining revised standard principles that consider the country’s unique circumstances.12 The authors recommended the following procedures: adherence to ATLS principles, administration of antibiotics, assessment for neurological and vascular injuries, immediate transfer of a threatened limb to a referral hospital, preliminary realignment and splinting, formal debridement only for gross contamination, no irrigation outside the operating theatre, debridement under anaesthesia, lavage with at least 5 litres of water before draping, photographic documentation, primary closure for clean wounds, fracture stabilization using external fixation or definitive fixation if appropriate soft tissue coverage is achieved, and amputation should only be performed for life-threatening injuries.12

    The purpose of this study was to conduct a narrative review of contemporary treatments for open tibial shaft fractures and assess their applicability to the South African context.

    Methods

    This study followed the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines13 and the updated recommendations provided in the Cochrane Handbook.14

    Eligibility Criteria

    This project incorporated all Level I–IV evidence-based clinical studies addressing open tibial shaft fractures. Reviews, systematic reviews, and meta-analyses were excluded from the analysis; however, their references were screened to identify relevant studies meeting inclusion criteria. Abstracts and conference proceedings were also excluded from the study.

    Literature Search

    A systematic review of the literature was performed to identify all publications in English and German, screening the databases Medline, Embase, Scopus, and Google Scholar. These databases were screened using the following terms and Boolean operators: “tibial fractures” AND/OR “open” AND/OR “compound” AND/OR “tibial shaft”; AND/OR “complications” AND/OR “treatment” AND/OR “management”. For the Medline search the MeSH term “tibia” was used with the following qualifiers: “fractures, bone” and “compound fractures” One reviewer conducted independent title and abstract screening. Disagreements between reviewers were resolved by consensus, and if no consensus was reached, they were carried forward to the full-text review. All eligible articles were manually cross-referenced to ensure that other potential studies were identified. The search period was restricted to studies published between 2000 and 2025 to ensure a contemporary review of current treatment approaches for open tibial shaft fractures.

    Data Extraction and Quality Assessment

    An electronic data extraction form was employed to systematically collect information from each article, including the level of evidence, study location, patient age, and sex. The key areas documented include incidence and epidemiology, fracture classification, treatment principles, antibiotic use, debridement, surgical timing, primary skin closure, temporary wound dressings, soft tissue management, large fragment management, and the applicability of these guidelines in resource-limited settings such as Southern Africa.

    Results

    Incidence – Epidemiology

    In the United Kingdom, open fractures of the lower extremity constitute approximately 12% of all open fractures, with an estimated incidence of 3.4 cases per 100,000 individuals annually.15 These injuries demonstrate a bimodal distribution, with high-energy trauma being the predominant cause in younger populations, while low-energy trauma is more common in older individuals, often attributable to decreased bone density.15 In the Netherlands, the estimated incidence of open fractures is approximately 1.1 per 100,000 person-years, with a notable increase observed in individuals over the age of 70.16 The German Trauma Registry reported nearly 3,000 open tibial fractures within a patient cohort of 148,000 over a ten-year period.17 Weiss et al reported an annual incidence of 2.3 per 100,000 person-years for open tibial fractures in Sweden and observed a decline in the overall incidence between 1998 and 2004.18 In a 15-year study analyzing 2,386 open fractures, the authors reported that 70% occurred in males, with only 22% resulting from road traffic accidents or falls from a height.15 However, when stratified, road traffic accidents accounted for 34% of lower extremity open fractures.15

    Unfortunately, there is a lack of comprehensive data published from the African continent. Existing studies only report the total number of treated cases, without providing information on the overall trauma burden or the proportion of open fractures among admitted trauma cases. For instance, Mwafulirwa et al reported that 72 open tibial fractures were managed at a tertiary hospital in Malawi during 2019.19 Almost all of these were caused by road traffic accidents (63%), assaults (18%), and falls (17%), with males accounting for the majority of cases (82%).19 Adesina et al reported similar findings, with motorcycle riders, artisans, and farmers accounting for 63% of open fractures, of which 75% occurred in male patients.20 While Clelland et al reviewed 1016 orthopaedic inpatients admitted in Northern Tanzania, their results demonstrated 143 had open tibia fractures.21

    Classification Systems

    The two most commonly utilized classification systems for open fractures are the Gustilo-Anderson scheme21,22 and that of the Orthopaedic Trauma Association (OTA). The Gustilo-Anderson system categorizes open fractures into three grades based on wound size, extent of skin loss, and muscle damage.21,22 Generally, Type I fractures involve a clean wound less than 1 cm in length, Type II fractures feature a laceration greater than 1 cm without significant soft tissue damage, flaps, or avulsions, and Type III fractures are characterized by open segmental fractures, extensive soft tissue damage, or associated vascular injury. This classification is widely accepted due to its ability to correlate severity grades with complication rates.23 However, it has been criticized for demonstrating poor to moderate inter-observer reliability.24

    The Orthopaedic Trauma Association (OTA) classification system evaluates open fractures based on five components: skin injury, muscle injury, arterial injury, contamination, and bone loss. Each component is assessed using predefined criteria and rated on a scale from 1 (mild) to 3 (severe).25,26 Compared to the Gustilo-Anderson classification, the Orthopaedic Trauma Association system demonstrated moderate to excellent inter-observer reliability. Additionally, it has been shown to outperform the Gustilo-Anderson system in predicting post-operative complications and clinical outcomes.27 The OTA classification system was recently updated to include a category for classifying post-traumatic bone defects.28

    Although the OTA open fracture classification is more complex, it is not necessarily superior to the Gustilo-Anderson system in predicting fracture-related infections.29 Given that the Gustilo-Anderson classification is easier to remember and more widely recognized, it may be the preferred choice for assessing fracture severity in resource-limited settings such as Southern Africa.

    Principles of Treatment

    Antibiotics

    Antibiotic prophylaxis is widely recognized for reducing infection rates and particularly in preventing early infections. Mundy et al provided comprehensive recommendations for antibiotic prophylaxis in open fractures.30 For Gustilo-Anderson Type I and Type II injuries, primary coverage against gram-positive organisms is advised, typically using a first-generation cephalosporin. Prophylaxis should not extend beyond 24 hours following wound closure.30 For Type III injuries, both gram-positive and gram-negative coverage is recommended. This is achieved with a combination of a first-generation cephalosporin and an aminoglycoside. Antibiotic administration should continue for 72 hours but should not exceed 24 hours after wound closure.30 In cases of farm injuries, additional anaerobic coverage is necessary, typically using penicillin. The same timelines for antibiotic administration as those outlined for Type I–III open fractures are applicable in these cases.28 The German guidelines recommend first- or second-generation cephalosporins for Type I–III fractures, with gram-negative coverage using ampicillin/sulbactam, piperacillin, or tazobactam.10 If Clostridia is suspected, penicillin or clindamycin should be added. Antibiotic administration should begin promptly, with a duration of no more than 24 hours for Type I and II fractures, and 72 hours for Type III fractures, but no longer than 24 hours post-wound closure.10 The Orthopaedic Trauma Association (OTA) recommends using cefazolin, clindamycin, or vancomycin for Type I and II fractures, with the addition of an aminoglycoside for Type III fractures. Alternatively, a combination of piperacillin and tazobactam is suggested for Type III fractures. Importantly, antibiotics should be administered within one hour of injury, and continued for no more than 24 hours for Type I and II injuries, and 72 hours for Type III injuries.31 The current AAOS guidelines on the prevention of surgical site infection after major extremity trauma give a moderate strength of recommendation for the administration of initial and preoperative antibiotics.32

    The only guidelines established through a consensus project have been published for Malawi.12 They recommend administering intravenous antibiotics as soon as possible, ideally within one hour of presentation.12 The guidelines suggest using ceftriaxone or a combination of doxycycline and gentamicin, with the addition of metronidazole for grossly contaminated wounds.12

    The use of local antibiotics remains unclear, though it offers the potential advantage of higher antibiotic concentrations compared to intravenous delivery.33 A meta-analysis has demonstrated a 12% reduction in risk with the use of local antibiotics.34 In a systematic review and meta-analysis, Craig et al demonstrated that the local administration of antibiotics significantly reduced the incidence of infection in Grade III fractures, from 31% to 9%.35 The VANCO trial, which administered vancomycin powder directly to the fracture site, reported a 6.4% probability of deep infection by 182 days in the treatment group, compared to 9.8% in the control group, suggesting promising results.36 Pesante and Parry demonstrated that the use of vancomycin and tobramycin powder reduced the rate of deep infections following open fracture treatment, thereby confirming the findings of the VANCO trial.37 The current AAOS guidelines strongly recommend the administration of local vancomycin powder or tobramycin-impregnated beads for the prevention of surgical site infection after major extremity trauma.32

    The timing of antibiotic administration appears to be a critical factor in preventing infection. Zuelzer et al demonstrated that administering antibiotics within 150 minutes of injury significantly reduces infection risk, even after adjusting for potential confounding factors such as age, diabetes, and smoking status.38 Earlier, Patzakis and Wilkins identified timely antibiotic administration as crucial in reducing infection risk.39 In their case-control study of over 1,100 open fractures, administering antibiotics more than three hours post-injury increased the odds of infection by 1.63 times compared to treatment within the first three hours.39 Extending the duration of antibiotic prophylaxis beyond 24 hours has not demonstrated a significant benefit in reducing the risk of fracture site infections.40 In contrast, the 2017 British Orthopaedic Association recommend administering antibiotics within one hour of injury, citing a 17% reduction in infection risk compared to those receiving antibiotics after 60 minutes.41

    Antibiotic-coated nails were first described by Paley and Herzenberg for the treatment of intramedullary infections.42 However, recent studies have highlighted their potential role in the primary treatment of open tibial fractures. A recent meta-analysis, which included only two studies, indicated a trend but no statistically significant differences toward reduced infection rates with the use of antibiotic-coated nails, identifying a 17% relative risk reduction in infection.43 Similarly, De Meo et al, in a systematic review of eight studies, found no evidence of advantages associated with antibiotic-coated nails in terms of fracture-related infection, non-union, or healing in both primary and revision surgeries.44 Given the current evidence, the use of an antibiotic-coated nail for primary fracture fixation in open tibial fractures cannot be recommended. Further high-quality randomized controlled trials are needed to clarify the potential benefits of this treatment option.

    Debridement

    Debridement involves thoroughly cleaning the wound by excising necrotic and devitalized tissue and removing foreign materials. It is a critical factor in achieving optimal outcomes in the management of open tibial fractures.22 Before surgical debridement, careful wound cleansing using a soft brush and a soap solution should be considered to reduce contamination.43 Careful excision of wound margins to healthy tissue is essential; however, undermining soft tissues and preserving tenuous skin bridges should be avoided to minimize the risk of compromised healing.10 Nonviable bone fragments should be removed, and any fragments that can be easily detached without resistance (using the “tug test”) should also be excised.10 Fragments that remain attached to the periosteum, however, should be preserved.10 Contaminated bone fragments should be thoroughly cleaned, debrided, and decorticated if necessary. The routine use of a tourniquet during these procedures is generally discouraged.10 If there is uncertainty about tissue viability, a second-look debridement should be considered, particularly in cases of small wounds with significant comminution, as the initial appearance may be misleading.3

    The traditional approach of routinely debriding all open tibial fractures within 6 hours no longer appears universally applicable. The current NICE guidelines recommend immediate debridement for wounds with vascular compromise, debridement of high-energy or contaminated wounds within 12 hours, and debridement of low-energy open fractures within 48 hours.39 These recommendations align with those of other established guidelines.10,31 Interestingly, studies suggest that factors other than time to debridement play a more significant role in perioperative infection risk. Independent risk factors include smoking, diabetes, prolonged surgical time, and fracture severity. Type III injuries, in particular, are associated with higher rates of reoperation and infection.46,47

    The Malawi guidelines recommend performing debridement in the operating room under general or spinal anaesthesia.12 They advise immediate debridement for highly contaminated wounds or cases wit vascular compromise, within 12 hours for Grade II and III fractures, and within 24 hours for Grade I fractures.12

    Irrigation

    Wound irrigation is a crucial component of open tibial fracture management, effectively removing contaminants and reducing the risk of infection. Current controversies focus on the debate between high-pressure versus low-pressure lavage and the selection of the optimal irrigation fluid.

    Studies suggest that high-pressure pulsatile lavage is more effective at removing bacteria and debris compared to low-pressure lavage.48 The main concern with high-pressure lavage is the potential to push contaminants deeper into tissues, which may increase infection rates and cause further damage to soft tissues and bone.49 The FLOW trial has provided clarity, demonstrating that warm normal saline with low-pressure irrigation should be the primary and safest choice for wound lavage.50 Conversely, Omar et al concluded that there is a lack of evidence to warrant discontinuing the use of pulsatile high-pressure lavage and recommended its continued implementation.10 Regarding irrigation volume, there is general agreement that 3 litres are sufficient for Type I injuries, 6 litres for Type II, and 9 litres for Type III injuries. However, it is generally accepted that highly contaminated wounds may require larger volumes until they are adequately cleansed of contamination.10,31,41,45 Irrigation fluids containing surfactants and antiseptics are no longer recommended, as they can cause secondary injury to the wound, increasing the risk of soft tissue necrosis.10,48 Furthermore, antimicrobial agents such as bacitracin have been shown to be associated with higher rates of wound healing complications.45 The AAOS guidelines strongly recommend irrigating wounds with saline without additives for initial wound management.32

    The Malawi guidelines advise against performing washouts outside the operating room and recommend that lavage be done in conjunction with debridement.12 They suggest using at least 5 litres of tap water followed by a minimum of 2 litres of sterile fluid.12

    Timing of Surgery and Surgical Implant Options

    The treatment of open tibial fractures should adhere to the general principles of orthopaedic trauma management, and the presence of an open fracture should not justify the departure from established osteosynthesis guidelines.10 Primary treatment is largely determined by the fracture characteristics, with both internal and external fixation techniques being viable options.10,51 An exception occurs when bone defects are present, which necessitate the use of appropriate reconstruction techniques.10 In general, Type I and II open fractures can be treated primarily with definitive osteosynthesis.10,52 Most Type III injuries can also follow this approach, except in cases with large or segmental bone defects, significant soft tissue damage requiring flap coverage, severely contaminated farm injuries, or cases involving vascular injuries that necessitate urgent vascular reperfusion surgery.10,52 For primary fixation, options include intramedullary fixation, plating, and external fixation methods such as ring fixators, hexapods, and static frames.10,52 Intramedullary nailing is generally considered the primary treatment option for most open diaphyseal and extra-articular metaphyseal fractures, although alternative fixation methods may be necessary in certain cases.31 Yokohama et al demonstrated that immediate reamed or unreamed nailing for Grade 3B and 3C fractures results in higher infection rates and should be avoided.53 However, the authors also concluded that other factors, such as early debridement, timely conversion of external fixation to nailing, and prompt skin closure, are critical in reducing the risk of deep infection.53 Intramedullary nailing can also be considered to be an effective bridging device for open fractures with bone loss.31 If definitive skeletal stabilization is not feasible for any reason, temporary spanning external fixation is an effective alternative.2,10,41 Temporary external fixation should be particularly considered in cases of severe contamination, extensive soft tissue involvement, or in unstable patients.3 Furthermore, the Ganga Hospital Open Injury Score (GHOIS) can aid in decision-making, with definitive fixation typically being appropriate when the score is below 9.54 The current AAOS guidelines on preventing surgical site infections after major extremity trauma provide a moderate-strength recommendation for definitive fracture fixation at the initial debridement, along with primary wound closure when appropriate. They also suggest that temporary external fixation remains a viable option.32

    The Malawi guidelines recommend that definitive internal stabilization should only be performed when it can be immediately followed by definitive soft tissue coverage.12 They also suggest that Grade IIIA and IIIB fractures be stabilized with an external fixator at the time of debridement.12 However, no specific recommendations were made regarding other surgical fixation methods.12

    Primary Skin Closure – Temporary Wound Dressings

    Historically, immediate primary closure of open fractures was thought to increase the risk of wound infection and fracture non-union.55 However, recent published literature has challenged this long-standing assumption. Hohmann et al reported no significant difference in infection rates between patients who underwent primary closure, with an average infection rate of 4%, and those who underwent delayed closure, which had an average infection rate of 2% when primary closure was performed.56 Moola et al demonstrated that primary closure for all open fractures is safe and does not increase the risk of postoperative infection.55 Their study identified no significant correlation between fracture classification, trauma velocity, or time to wound closure and the occurrence of infection, delayed union, or non-union.50 Scharfenberger et al demonstrated that primary wound closure in Grade I–IIIA open fractures resulted in lower rates of infection (4% vs 9%) and nonunion (13% vs 29%) compared to delayed closure.57 Rajasekaran reported that primary wound closure is safe when performed under specific conditions: debridement is completed within 12 hours, there is no significant skin loss, skin approximation is achievable without tension, and there is no evidence of vascular insufficiency.54 Riechelmann et al confirmed that primary soft tissue closure is safe for Grade I–IIIA open fractures, provided that debridement is thorough, the skin margins are bleeding and viable, and appropriate antibiotics are administered.58 It is noteworthy, and perhaps counterintuitive, that re-exploration of the wound during definitive fracture fixation does not appear to be associated with an increased risk of complications.59 Reynolds et al reported no significant difference in complication rates between patients with open tibia fractures who underwent staged fixation.59

    Primary closure is generally recommended for Type I to Type IIIA tibial fractures when sufficient viable soft tissue is available to achieve tension-free closure. This approach is contingent on meticulous debridement of the injury and the timely administration of prophylactic antibiotics.60 The current AAOS guidelines on preventing surgical site infections after major extremity trauma strongly recommend the use of negative pressure therapy, as it may reduce the risk of revision surgery and superficial site infections.32 However, silver-coated dressings are generally not recommended, with only a moderate-strength recommendation.32 Regarding primary wound closure, the guidelines strongly recommend closure when feasible and when there is no significant gross contamination.32

    The Malawi guidelines recommend primary closure for clean Grade I fractures, leaving Grade II fractures open with closure within 72 hours, and keeping Grade III fractures open.12 For Grade III fractures, patients should be referred to the nearest specialized hospital for further management.12

    Soft Tissue Management

    For fracture wounds that cannot be closed primarily and may require flap coverage, the injury location, defect size, and zone of damage must be carefully assessed to determine whether rotational or free flap coverage is the most suitable option.30 Fractures in the proximal two-thirds of the tibia are typically treated with rotational muscle flaps, while those in the distal third generally require free flaps.30 Soft tissue management should aim to achieve flap coverage within 72 hours to minimize the risk of deep infection.10 Lack et al reported that delaying soft tissue closure beyond 5 days doubles the infection rate.61

    In cases where primary wound closure is not possible and temporary wound management is needed, negative pressure wound therapy is an effective option.10 Kim and Lee demonstrated in a meta-analysis that negative pressure wound therapy, compared to conventional management, resulted in lower rates of soft tissue infections, non-union, flap necrosis, and the need for revisions.62 Stannard et al reported in a randomized controlled trial that negative pressure wound therapy significantly reduced the total infection rate (acute and late combined) compared to saline-soaked dressings, although the estimate lacked precision.63 In a similar study, Kumaar et al demonstrated that negative pressure wound therapy significantly reduced infections and enhanced the healing of open fracture wounds.64 However, both the WHIST and WOLLF trials found no evidence that negative pressure wound therapy (NPWT) reduced infection rates compared to open solid foam or gauze dressings.65,66 However, the WOLLF trial was conducted in the UK, and all open fractures in their cohort underwent definitive soft tissue management within 72 hrs from injury, perhaps negating any benefit NPWT may have provided. Regardless of the wound management method, five-year results from the WHIST trial still reported high levels of persistent disability and reduced quality of life, with minimal evidence of improvement over this period.67 The current AAOS guidelines on preventing surgical site infections after major extremity trauma provide a moderate-strength recommendation for wound closure within seven days.30 However, they note that the current evidence supporting the use of an orthoplastic team or hyperbaric oxygen therapy is limited.32

    Large Bone Fragments

    The presence of large bone fragments, whether devitalized, extruded, or attached to viable soft tissue, remains a significant challenge and a subject of ongoing debate.68 Traditionally, the standard approach has been to discard devitalized or extruded cortical fragments; however, this practice has recently been questioned. In cases of severe contamination or comminution, such as ballistic injuries, retaining bone fragments is not feasible, and discarding them may be the most logical and often only option for the surgeon.68 If large bone fragments remain attached to the periosteum or pass the tug test (showing substantial resistance when attempting to remove them), they may be preserved and reduced if possible.10 Devitalized and extruded fragments can be retained if thoroughly debrided and disinfected to reduce bacterial load before being incorporated into the fracture site.69 Mechanical scrubbing followed by a five-minute immersion in povidone-iodine or chlorhexidine appears to be a safe and effective time interval.70 Another author has suggested soaking the fragment in a vancomycin solution for an additional thirty minutes to further reduce the risk of infection.71 The Bristol experience demonstrated that incorporating mechanically relevant, debrided devitalized bone fragments into the definitive reconstruction of Type IIIB open diaphyseal tibial fractures is a safe approach.69 In addition, two case reports demonstrated the successful reimplantation of extruded bone fragments.71,72

    Conclusions

    The initial treatment of open tibial fractures remains controversial and lacks robust recommendations. Key steps include early administration of intravenous antibiotics, timely debridement and lavage of open wounds, primary wound closure when tissue is viable and closure can be achieved without tension, and early flap coverage within 72 hours if needed. Preferred definitive stabilization for Grade I–IIIa fractures is intra-medullary nailing, with temporary external fixation used when necessary. Early conversion to definitive treatment is also essential.

    In low-resource countries in Southern Africa, only one guideline has been developed, which recommends the administration of early intravenous antibiotics, timely debridement and irrigation in the operating room, and management based on fracture severity. The guideline advises primary closure for Grade 1 fractures, delayed closure for Grade 2 fractures, and no closure for Grade 3 fractures, with referral to a specialist hospital for further management of Grade 3 injuries. In this context, further exploration is needed regarding the applicability of early simple oral antibiotics as an alternative to intravenous administration, the use of locally administered antibiotics, and temporary fixation with homemade antibiotic nails. Furthermore, optimal timing for both initial and definitive surgery, the use of temporary or permanent wound dressings, and soft tissue management when referral is not possible or significantly delayed require further investigation. The management of large bone fragments at the time of debridement also warrants further investigation.

    Ultimately, the absence of general recommendations and context-specific guidelines for the initial management of open tibial fractures in Southern Africa highlights the need for further work. Specific issues to address include how to evaluate and treat these injuries in low-resource settings that are by staff shortages, inadequate infrastructure, and inconsistent medication supply.

    Author Contributions

    All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis, and interpretation, or in all these areas; took part in drafting, revising, or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

    Funding

    This research did not receive any funding.

    Disclosure

    Professor Kevin Tetsworth is an unpaid consultant for AO Foundation, personal fees for speakers bureau and design consultant from Smith and Nephew, personal fees for speakers bureau from Johnson and Johnson MedTech, scientific advisory board for and shares and stock options from OrthoDx and VitaClot Medical, outside the submitted work. The authors report no other conflicts of interest in this work.

    References

    1. Qian S, Shen Y, Sun L, Wang Z. Treatment preferences and current practices regarding open tibial fractures. Front Public Health. 2024;12:1331654. doi:10.3389/fpubh.2024.1331654

    2. Nicolaides M, Pafitanis G, Vris A. Open tibial fractures: an overview. J Clin Orthop Trauma. 2021;20:101483. doi:10.3389/fpubh.2024.1331654

    3. Dheenadhaylan J, Nagashree V, Devendra A, Velmurugesan PS, Rajasekaran S. Management of open fractures: a narrative review. J Clin Orthop Trauma. 2023;44:102246. doi:10.1016/j.jcot.2023.102246

    4. Ataguba JE, McIntyre D. Paying for and receiving benefits from health services in South Africa: is the health system equitable? Health Policy Plan. 2012;27(1):i35–45. doi:10.1093/heapol/czs005

    5. Coovadia H, Jewkes R, Barron P, Sanders D, McIntyre D. The health and health system of South Africa: historical roots of current public health challenges. Lancet. 2009;374(9692):817–834. doi:10.1016/S0140-6736(09)60951-X

    6. Zubair AA, Abdullateef R, Davis S, et al. A scoping review on the management of open fractures in African trauma and orthopaedics centres. Cureus. 2024;16(9):368925. doi:10.7759/cureus.68925

    7. Schade AT, Sabawo M, Nyamulani N, et al. Functional outcomes and quality of life at 1-year follow-up after an open tibia fracture in Malawi: a multicentre, prospective cohort study. Lancet Global Health. 2023;11(10):e1609–e1618. doi:10.1016/S2214-109X(23)00346-7

    8. Shafiq B, Hacquebord J, Wright DJ, Gupta R. Modern principles of acute surgical management of open tibial distal fractures. J Am Acad Orthop Surg. 2021;29(11):e536–e547. doi:10.5435/JAAOS-D-20-00502

    9. Hohmann E, Birkholtz F, Glatt V, Tetsworth K. The “road to union” protocol for the reconstruction of isolated complex high-energy tibial trauma. Injury. 2017;48(6):1211–1216. doi:10.1016/j.injury.2017.03.018

    10. Omar M, Zeckey C, Krettek C, Graulich T. Open fractures. Unfallchirurg. 2021;124(8):651–665. doi:10.1007/s00113-021-01042-2

    11. Castillo IA, Heiner JA, Meremikwu RI, Kellam J, Warner SJ. Where are we in 2022? A summary of 11,000 open tibia fractures over 4 decades. J Orthop Trauma. 2023;37(8):e326–e334. doi:10.1097/BOT.0000000000002602

    12. Schade AT, Yesaya M, Bates J, Martin Jr C, Harrison WJ. The Malawi Orthopaedic Association/AO alliance guidelines and standards for open fracture management in Malawi: a national consensus statement. Malawi Med J. 2020;32(3):112–118. doi:10.4314/mmj.v32i3.2

    13. Moher D, Shamseer L, Clarke M, et al. Preferred reporting items for systematic reviews and meta-analysis protocols (PRISMA-P) 2015 statement. Syst Rev. 2015;4(1):1. doi:10.1186/2046-4053-4-1

    14. Cumpston M, Li T, Page MJ, et al. Updated guidance for trusted symptomatic reviews of the Cochrane handbook for systematic reviews of interventions. Cochrane Database Syst Rev. 2019;10:ED000142. doi:10.1002/14651858.ED000142

    15. Court-Brown CM, Burgler KE, Clement ND, Duckworth AD, McQueen MM. The epidemiology of open fractures in adults. A 15-year review. Injury. 2012;43(6):891–897. doi:10.1016/j.injury.2011.12.007

    16. Noorlander-Borgdorff MP, Sekercan A, Young-Afat DA, Bouman M, Botman M, Giannakopoulos GF. Nationwide study on open tibial fractures in the Netherlands: incidence, demographics and level of hospital care. Injury. 2024;55(6):111487. doi:10.1016/j.injury.2024.111487

    17. Weber CD, Hildebrand F, Kobbe P, et al. Epidemiology of open tibia fractures in a population-based database: update on current risk factors and clinical implications. Eur J Trauma Emerg Surg. 2019;45(3):445–453. doi:10.1007/s00068-018-0916-9

    18. Weiss RJ, Montgomery SM, Ehlin A, Al Dabbagh Z, Stark A, Jansson K-A. Decreasing incidence of tibial shaft fractures between 1998 and 2004: information based on 10,627 Swedish inpatients. Acta Orthop. 2008;79(4):526–533. doi:10.1080/17453670710015535

    19. Mwafulirwa K, Munthali R, Ghosten I, Schade A. Epidemiology of open tibia fractures presenting to a tertiary hospital in Southern Malawi: a retrospective study. Malawi Med J. 2022;34(2):118–122. doi:10.4314/mmj.v34i2.7

    20. Adesina SA, Amole IO, Owolabi JI, Oyewusi OO, Adefokun IG, Eyesan SU. Locked intramedullary nailing of open fractures in resource-poor settings: a prospective observational study of challenges and functional outcomes in 101 fractures at Ogbomoso, Nigeria. BMC Musculoskelet Disord. 2023;24(1):170. doi:10.1186/s12891-023-06271-7

    21. Clelland SJ, Chauhan P, Mandari FN. The epidemiology and management of tibia and fibula fractures at Kilimanjaro Christian Medical Centre (KCMC) in Northern Tanzania. Pan Afr Med J. 2016;25:51. doi:10.11604/pamj.2016.25.51.10612

    22. Gustilo RB, Anderson JT. Prevention of infection in the treatment of one thousand and twenty-five open fractures of long bones: retrospective and prospective analyses. J Bone Joint Surg Am. 1976;58(4):453–458.

    23. Gustilo RB, Mendoza RM, Williams DN. Problems in the management of type III (severe) open fractures: a new classification of type III open fractures. J Trauma. 1984;24(8):742–746. doi:10.1097/00005373-198408000-00009

    24. Thakore RV, Francois EL, Nwosu SK, et al. The Gustilo-Anderson classification system as predictor of nonunion and infection in open tibia fractures. Eur J Trauma Emerg Surg. 2017;43(5):651–656. doi:10.1007/s00068-016-0725-y

    25. Brumback RJ, Jones AL. Interobserver agreement the classification of open fractures of the tibia. The results of a survey of two hundred and forty-five orthopaedic surgeons. J Bone Joint Am. 1994;76(8):1162–1166. doi:10.2106/00004623-199408000-00006

    26. Orthopaedic Trauma Association: Open Fracture Study Group. A new classification system scheme for open fractures. J Orthop Trauma. https://ota.org/sites/files/2021-06/General%20B2%20Open%20Fractures.pdf. 2010;24(8):457–464. doi:10.1097/BOT.0b013e3181c7cb6b

    27. Hao J, Cuellar DO, Herbert B, et al. oes the OTA Open Fracture Classification Predict the Need for Limb Amputation? A Retrospective Observational Cohort Study on 512 Patients. J Orthop Trauma. 2016;30(4):194–198. doi:10.1097/BOT.0000000000000479

    28. Tetsworth KD, Burnand HG, Hohmann E, Glatt V. Classification of Bone Defects: An Extension of the Orthopaedic Trauma Association Open Fracture Classification. J Orthop Trauma. 2021;35(2):71–76. doi:10.1097/BOT.0000000000001896

    29. Khoury P, Hazra N, DeMartino A, et al. Is the Orthopaedic Trauma Association-Open Fracture Classification Better Than the Gustilo-Anderson Classification at Predicting Fracture-Related Infections in the Tibia? J Orthop Trauma. 2024;38(1):655–660. doi:10.1097/BOT.0000000000002907

    30. Mundy R, Chaudry H, Niroopan G, Petrisor B, Bhandari M. Open tibial fractures: updated guidelines for management. JBJS Rev. 2015;3(2):e1. doi:10.2106/JBJS.RVW.N.00051

    31. Orthopaedic Trauma Association. Open Fractures. https://ota.org/sites/files/2021-06/General%20B2%20Open%20Fractures.pdf. Accessed 7, Jan 2025.

    32. Goldman AH, Tetsworth K. AAOS Clinical Practice Guideline Summary: prevention of Surgical Site Infection After Major Extremity Trauma. J Am Acad Orthop Surg. 2023;31(1):e1–e8. doi:10.5435/JAAOS-D-22-00792

    33. Flores MJ, Brown KE, Morshed S, Shearer DW. Evidence for Local Antibiotics in the Prevention of Infection in Orthopaedic Trauma. J Clin Med. 2022;11(24):7461. doi:10.3390/jcm11247461

    34. Morgenstern M, Vallejo A, McNally MA, et al. The effect of local antibiotic prophylaxis when treating open limb fractures: a systematic review and meta-analysis. Bone Joint Res. 2018;7(7):447–456. doi:10.1302/2046-3758.77.BJR-2018-0043.R1

    35. Craig J, Fuchs T, Jenks M, et al. Systematic review and meta-analysis of the additional benefit of local prophylactic antibiotic therapy for infection rates in open tibia fractures treated with intramedullary nailing. Int Orth. 2014;38(5):1025–1030. doi:10.1007/s00264-014-2293-2

    36. Major Extremity Trauma Research Consortium (METRC), et al. Effect of Intrawound Vancomycin Powder in Operatively Treated High-risk Tibia Fractures: a Randomized Clinical Trial. JAMA Surg. 2021;156(1):e207259. doi:10.1001/jamasurg.2020.7259

    37. Pesante BD, Parry JA. The Effect of Vancomycin and Tobramycin Local Antibiotic Powder on Surgical Site Infections After Open Treatment of Fracture: a Retrospective Propensity-Matched Analysis. J Orthop Trauma. 2024;38(4):177–182. doi:10.1097/BOT.0000000000002758

    38. Zuelzer DA, Hayes CB, Hautala GS, Akbar A, Mayer RR, Jacobs CA. Early Antibiotic Administration Is Associated with a Reduced Infection Risk When Combined with Primary Wound Closure in Patients with Open Tibia Fractures. Clin Orth Relat Res. 2021;479(3):613–619. doi:10.1097/CORR.0000000000001507

    39. Patzakis MJ, Wilkins J. Factors influencing infection rate in open fracture wounds. Clin Orthop Relat Res. 1989;243:36–40.

    40. Dellinger EP, Caplan ES, Weaver LD, et al. Duration of preventive antibiotic administration for open extremity fractures. Arch Surg. 1988;123(3):333–339. doi:10.1001/archsurg.1988.01400270067010

    41. British Orthopaedic Association Trauma Committee. British Association Standard for Trauma (BOAST): open fracture management Injury. 2020;51(2):174–177. doi:10.1016/j.injury.2020.06.005.

    42. Paley D, Herzenberg JE. Intramedullary infections treated with antibiotic cement rods: preliminary results in nine cases. J Orthop Trauma. 2002;16:723–729. doi:10.1097/00005131-200211000-00007

    43. Vargas-Hernandez JS, Sanchez CA, Renza S, Leal JA. Effectiveness of antibiotic-coated intramedullary nails for open tibia fracture infection prevention. A systematic review and meta-analysis. Injury. 2023;54(6):110857. doi:10.1016/j.injury.2023.110857

    44. De Meo D, Cannari FM, Petriello L, Persiani P, Villani C. Gentamicin-Coated Tibia Nail in Fractures and Nonunion to Reduce Fracture-Related Infections: a Systematic Review. Molecules. 2020;25(22):5471. doi:10.3390/molecules25225471

    45. Anglen JO. Wound irrigation in musculoskeletal injury. J Am Acad Orthop Surg. 2001;9(4):219–226. doi:10.5435/00124635-200107000-00001

    46. Heckmann ND, Davis JA, Mombell K, et al. Delayed debridement of open tibia fractures beyond 24 and 48 h does not appear to increase infection and reoperation risk. Europ J Orthop Surg Traumatol. 2022;32(5):953–958. doi:10.1007/s00590-021-03057-2

    47. Li J, Wang Q, Lu Y, et al. Relationship Between Time to Surgical Debridement and the Incidence of Infection in Patients with Open Tibial Fractures. Orthop Surg. 2020;12(2):524–532. doi:10.1111/os.12653

    48. Elniel AR, Giannoudis PV. Open fractures of the lower extremity: current management and clinical outcomes. EFFORT Open Rev. 2018;3(5):316–325. doi:10.1302/2058-5241.3.170072

    49. Kortram K, Bezstarosti H, Metsemakers WJ, Raschke MJ, Van Lieshout EMM, Verhofstad MHJ. Risk factors for infectious complications after open fractures; a systematic review and meta-analysis. Int Orth. 2017;41(10):1965–1982. doi:10.1007/s00264-017-3556-5

    50. Flow Investigators, Bhandari M, Jeray KJ, Petrisor BA, Devereaux PJ, Heels-Ansdell D. A Trial of Wound Irrigation in the Initial Management of Open Fracture Wounds. N Engl J Med. 2015;373(27):2629–2641. doi:10.1056/NEJMoa1508502

    51. Crowley DJ, Kanakaris NK, Giannoudis PV. Irrigation of the wounds in open fractures. J Bone Joint Br. 2007;89(5):580–585. doi:10.1302/0301-620X.89B5.19286

    52. Duyos OA, Beaton-Comulada D, Davila-Parilla A, et al. Management of Open Tibial Shaft Fractures: does the Timing of Surgery Affect Outcomes. J Am Acad Orthop Surg. 2017;25(3):230–238. doi:10.5435/JAAOS-D-16-00127

    53. Yokohama K, Itoman M, Uchino M, Fukushima K, Nitta H, Kojima Y. Immediate versus delayed intramedullary nailing for open fractures of the tibial shaft: a multivariate analysis of factors affecting deep infection and fracture healing. Indian J Orthop. 2008;42(4):410–419. doi:10.4103/0019-5413.43385

    54. Rajasekaran S. Early versus delayed closure of open fractures. Injury. 2007;38(8):890–895. doi:10.1016/j.injury.2007.01.013

    55. Moola FO, Carli A, Berry GK, Reindl R, Jacks D, Harvey EJ. Attempting primary closure for all open fractures: the effectiveness of an institutional protocol. Can J Surg. 2014;57(3):E82–88. doi:10.1503/cjs.011413

    56. Hohmann E, Tetsworth K, Radziejowski MJ, Wiesnieswki TF. Comparison of delayed and primary wound closure in the treatment of open tibial fractures. Arch Orthop Trauma Surg. 2007;127(2):131–136. doi:10.1007/s00402-006-0222-6

    57. Scharfenberger AV, Alabassi K, Smith S, et al. Primary Wound Closure After Open Fracture: a Prospective Cohort Study Examining Nonunion and Deep Infection. J Orthop Trauma. 2017;31(3):121–126. doi:10.1097/BOT.0000000000000751

    58. Riechelmann F, Kaiser P, Arora R. Primary soft tissue management in open fracture. Oper Orthop Traumatol. 2018;30(5):294–308. doi:10.1007/s00064-018-0562-8

    59. Reynolds AW, Garay M, Philp FH, Hammarstedt, Altman GT, Nwankwo CH. Definitive fixation of open tibia fractures: does reopening the traumatic wound increase complication rates? J Clin Orthop Trauma. 2021;24:101715. doi:10.1016/j.jcot.2021.101715

    60. Melvin JS, Dombroski DG, Torbert JT, Kovach SJ, Esterhai JL, Mehta S. Open tibial shaft fractures: i. Evaluation and initial wound management. J Am Acad Orthop Surg. 2010;18(1):10–19. doi:10.5435/00124635-201001000-00003

    61. Lack WD, Karunakar MA, Angerame MR, et al. Type III open tibia fractures: immediate antibiotic prophylaxis minimizes infection. J Orthop Trauma. 2015;29(1):1–6. doi:10.1097/BOT.0000000000000262

    62. Kim JH, Lee DH. Negative pressure wound therapy vs. conventional management in open tibia fractures: systematic review and meta-analysis. Injury. 2019;50(10):1764–1772. doi:10.1016/j.injury.2019.04.018

    63. Stannard JP, Volgas DA, Stewart R, McGwin G, Alonso JE. Negative pressure wound therapy after severe open fractures: a prospective randomized study. J Orthop Trauma. 2009;23(8):552–557. doi:10.1097/BOT.0b013e3181a2e2b6

    64. Kumaar A, Shanhappa AH, Ethiraj P. A Comparative Study on Efficacy of Negative Pressure Wound Therapy Versus Standard Wound Therapy for Patients With Compound Fractures in a Tertiary Care Hospital. Cureus. 2022;14(4):e23727. doi:10.7759/cureus.2372

    65. Costa ML, Achten J, Bruce J, et al. UK WOLLF Collaboration. Effect of Negative Pressure Wound Therapy vs Standard Wound Management on 12-Month Disability Among Adults With Severe Open Fracture of the Lower Limb: the WOLLF Randomized Clinical Trial. JAMA. 2018;319(22):2280–2288. doi:10.1001/jama.2018.6452

    66. Costa ML, Achten J, Knight R, Png ME, Bruce J, Dutton S. Negative-pressure wound therapy compared with standard dressings following surgical treatment of major trauma to the lower limb: the WHiST RCT. Health Technol Assess. 2020;38(1):1–86. doi:10.3310/hta24380

    67. Costa ML, Achten J, Knight R, Campolier M, MS M. Five-year outcomes for patients sustaining severe fractures of the lower limb from the Wound Healing in Surgery for Trauma (WHIST) trial. Bone Joint J. 2024;106(8):858–864. doi:10.1302/0301-620X.106B8

    68. Farhan-Alanie M, Ward J, Kelly MB, Al-Hourani K. Current Perspectives on the Management of Bone Fragments in Open Tibial Fractures: new Developments and Future Directions. Orthop Res Rev. 2022;14:275–286. doi:10.2147/ORR.S340534

    69. Al-Hourani K, Stoddard M, Khan U, Riddick A, Kelly M. Orthoplastic reconstruction of type IIIB open tibial fractures retaining debrided devitalized cortical segments: the Bristol experience 2014 to 2018. Bone Joint J. 2019;101(8):1002–1008. doi:10.1302/0301-620X.101B8.BJJ

    70. Bruce B, Sheibani-Rad S, Appleyard D, et al. Are dropped osteoarticular bone fragments safely reimplantable in vivo? J Bone Joint Surg Am. 2011;93(5):430–438. doi:10.2106/JBJS.J.00793

    71. Shanmuganathan R, Chandra Mohan AK, Agraharam D, Perumal R, Jayaramaraju D, Kulkarni S. Successful reimplantation of extruded long bone segments in open fractures of lower limb–a report of 3 cases. Injury. 2015;46(7):1389–1392. doi:10.1016/j.injury.2015.04.006

    72. Rathore S, Reddy IV, Kumar AHA. A novel technique for reimplanting extruded bone fragments in open fractures. Trauma Case Rep. 2016;4:5. doi:10.1016/j.tcr.2016.05.006

    Continue Reading

  • MVA-BN Vaccine Still Protects Against Severe Mpox, But Boosters and Equity Challenges Remain

    MVA-BN Vaccine Still Protects Against Severe Mpox, But Boosters and Equity Challenges Remain

    A new retrospective cohort study published in The Lancet Primary Care has provided compelling real-world evidence that the MVA-BN (Modified Vaccinia Ankara-Bavarian Nordic) vaccine continues to offer substantial protection against severe monkeypox (mpox) disease more than 1.5 years after immunization. The commentary accompanying the study—written by infectious disease experts Dr. Emily Evans and Dr. Boghuma Titanji of Emory University—highlights how the findings mark a significant step forward in understanding vaccine durability and clinical protection in the face of waning antibody levels.

    The original research, conducted in New South Wales (NSW), Australia, by Latham et al., represents the largest and most detailed clinical study to date on breakthrough mpox infections in fully vaccinated individuals. It is particularly significant given the limited baseline immunity in the Australian population due to the absence of historical smallpox vaccination campaigns, a contrast to settings in Europe, North America, and parts of Africa.

    Durable Protection Despite Waning Antibodies

    The study included 674 confirmed mpox cases between June and November 2024, during Australia’s largest recorded mpox outbreak. Of those, 251 individuals (37%) were fully vaccinated with two doses of MVA-BN. The median interval between the second dose and symptom onset was nearly 22 months.

    Key outcomes demonstrate that full vaccination was associated with:

    • 89% reduced risk of hospitalization compared to unvaccinated individuals (RR 0.11; 95% CI 0.03–0.43),
    • 55% reduction in risk of systemic symptoms like fever, headache, and muscle pain (RR 0.72),
    • Significant reduction in extragenital lesions, a potential driver of casual-contact transmission (RR 0.45).

    Interestingly, fully vaccinated individuals were slightly more likely to present with anogenital lesions, though these were generally mild and did not lead to hospitalization.

    Despite laboratory evidence that antibody titers wane within 5–7 months of vaccination, these clinical findings suggest meaningful protection persists well beyond serological decline. This divergence between antibody kinetics and real-world outcomes adds nuance to the discussion around correlates of protection and vaccine-induced immunity.

    Implications for HIV-Positive Individuals

    While the Australian cohort included people living with HIV, the study did not stratify clinical outcomes by HIV status. This is a notable omission, given that HIV-positive individuals—especially those with advanced disease—are at substantially higher risk of severe mpox, including disseminated and even fatal outcomes.

    Previous research has shown that while people with HIV can mount adequate immune responses to MVA-BN, those with lower CD4 counts or poorly managed HIV may have weaker protection. In one clinical trial, antibody titers were significantly higher in people without HIV compared to those with HIV, despite both groups reaching high seropositivity rates.

    The absence of outcome data stratified by HIV status represents a critical gap, especially as current mpox outbreaks in sub-Saharan Africa—where HIV prevalence is high—continue to intensify.

    A Public Health Opportunity and National Imperative

    The findings have important implications for both national and global health security. Infections with mpox, while relatively rare outside high-risk populations, can escalate quickly during outbreaks and impose significant strain on healthcare systems.

    Preventing hospitalizations and systemic symptoms—especially in younger, unvaccinated populations—is not only a clinical goal but also a matter of public health resilience. In the Australian outbreak, the vast majority of mpox patients were men under 50, underscoring the vulnerability of generations never immunized against smallpox.

    As Dr. Titanji and Dr. Evans note, leveraging sexual health clinics as vaccination hubs has proven effective, but disparities in access persist. Populations disconnected from such services may face increased risk during future outbreaks, further emphasizing the need for broader, community-engaged vaccination strategies.

    Global Equity and the Next Phase: Boosters?

    The NSW study’s implication that protection extends well beyond the initial year is reassuring—but it also raises new questions. Should a third (booster) dose be recommended for specific populations? Should HIV-positive individuals, or those with high behavioral exposure risk, receive tailored vaccine schedules?

    The commentary urges global immunization technical advisory groups to incorporate these findings into deliberations about MVA-BN booster strategies. It also reinforces the call for equitable global access to MVA-BN, especially in lower-income settings currently grappling with resurging outbreaks.

    Notably, viral sequencing in the Australian outbreak revealed no substantial genetic divergence between viruses infecting vaccinated and unvaccinated individuals. This suggests vaccine escape was not a major factor in breakthrough infections and bolsters confidence in the ongoing relevance of the MVA-BN platform.

    Next Steps: Research, Policy:

    This evidence base provides a firm foundation for future preparedness—but several priorities remain:

    • Stratified clinical studies on vaccine efficacy in immunocompromised populations,
    • Longitudinal analyses of immune response durability post-MVA-BN,
    • Global coordination on vaccination campaigns in areas with rising case counts and high HIV prevalence,
    • Booster dose policy frameworks informed by risk profiling, not just time since vaccination.

    As mpox transitions from a global health emergency to an endemic risk, the world must shift from reactive containment to proactive, equitable protection—particularly for communities historically underserved by immunization programs.


    Sources and Further Reading

    Evans EE, Titanji BK. Protection that lasts? MVA-BN against clade IIb mpox. The Lancet Primary Care, 24 July 2025.

    Latham NH, Pett J, Katelaris AL, et al.  Clinical features of mpox in fully vaccinated people in New South Wales, Australia: a retrospective cohort study. The Lancet Primary Care, 24 July 2025.

    Taha AM, Rodriguez-Morales AJ, Sah R. Mpox breakthrough infections: concerns and actions. The Lancet Infectious Diseases, Nov 2023.

    Valentina M, Guiulia M, Eleonora C, et al. Humoral and T-Cell Responses Following MVA-BN Booster Vaccination Against Mpox Virus Clades Ib and IIb,  MedRxiv pre-print, 7 July 2025.

    Continue Reading

  • Polaris Genomics Secures $250,000 Debt in Quest to Identify Biomarkers for Mental Health Disorders

    Polaris Genomics Secures $250,000 Debt in Quest to Identify Biomarkers for Mental Health Disorders

    Gaithersburg, Md.-based Polaris Genomics, a mental healthcare firm eyeing physical biomarkers to aid diagnoses as well as treatments, has secured $250,000 in debt, according to a recent SEC filing. The company raised over $5.9 million from investors, according to Pitchbook.

    The startup up was founded in 2017 by Yusuf Henriques, a US Army veteran, Charles Cathlin, who served in the US Air Force, Princeton graduate Tshaka Cunningham and physician Anne Naclerio, a 30-year veteran of the US Army, with Cathlin drawing on his experience treating the first responders of the 9/11 terror attacks of 2001.

    Cathlin serves as the company’s CEO and Cunningham is chief science officer. Cunningham is director of R&D while Naclerio serves as chief medical officer.

    According to a report in Startup Health, Cathlin — an engineer and graduate of the Air Force Academy and Stanford University — grappled with the unique clinical challenge of “diagnosing” mental health conditions, rather than relying on observation or self-reporting.

    “The firefighters and police officers were traumatized by what they experienced,” Cathlin told Illumina, whose accelerator Polaris Genomics joined as an early-stage startup. “That had a large impact on me and got me interested in mental health, because no one was really addressing it.”

    In his quest, Cathlin eventually tracked down the work of a psychiatric researcher in New York City that connected mental health conditions and RNS expression.

    “What got me excited was that there was an actual biomarker to identify who actually had post-traumatic stress disorder,” Cathlin told Startup Health. “The fact that you can introduce an objective tool to that process was a game changer.”

    Cathlin believes the company’s mission is to make “invisible wounds visible using the power of genomics.”

    “We believe genomics is a driving force to connect mental illness to its biological underpinnings. By doing that, we can decrease the stigma, suffering, silence, and suicide.”

    Polaris Genomics has partnered with the Illumina Accelerator, and scientists from Mount Sinai and the Max Planck Institute of Psychiatry to develop the first-to-market, patented genomic biomarker assay to identify risks of post-traumatic stress disorder (PTSD). The company was granted two patents on the blood-based biomarker model for PTSD.

    Last year, the company launched a trial with Stella DC to further develop PTS-iD, called a first test of its kind to use genomics to detect the physical changes underlying PTSD. Polaris Genomics believes the Covid-19 pandemic and subsequent years of mental health repercussions has “elucidated the urgency for advances in diagnostics and treatment across a range of mental health and neuropsychiatric conditions.”

    Continue Reading

  • The Role of Plasma Metabolites in Mediating the Effect of Gut Microbio

    The Role of Plasma Metabolites in Mediating the Effect of Gut Microbio

    Background

    Obstructive sleep apnea (OSA) is a common sleep disorder, with a prevalence of approximately 23% in women and nearly 50% in men.1 The development of OSA is influenced by both anatomical and non-anatomical risk factors. Anatomical factors, such as obesity and craniofacial abnormalities, promote upper airway collapse and obstruction during sleep.2 Non-anatomical factors include heightened airway collapsibility, a low respiratory arousal threshold, and unstable ventilatory control.3 The hallmark feature of OSA is intermittent hypoxia during sleep, which significantly increases the risk of cardiovascular, neurological, and metabolic disorders.4

    The potential role of the gut microbiota in human disease development has received increasing attention over the past decade. In particular, microbiota dysbiosis has been implicated in the pathogenesis of OSA.5 Disruption of the gut microbiota has been associated with altered sleep-wake architecture6 and reduced sleep efficiency.7,8 It may also impair the ventilatory response to hypercapnia,9 thereby contributing to unstable ventilatory control in patients with OSA. In addition, microbiota dysbiosis contributes to the development of obesity,10 a key risk factor for OSA. Probiotics have emerged as promising adjunctive interventions for OSA, primarily through their ability to reduce systemic inflammation and enhance gut barrier integrity.11 Faecal microbiota transplantation has also been shown to partially alleviate cardiovascular disturbances induced by chronic intermittent hypoxia.12 Moreover, the probiotic strain Lactobacillus rhamnosus GG has demonstrated beneficial effects in experimental models of OSA, including improvements in metabolic disturbances,13 mitigation of hypertension severity,14 attenuation of cardiac remodeling and inflammation.15 Accordingly, elucidating the role of gut microbiota in the pathogenesis of OSA holds substantial promise, and the identification of effective probiotic-based interventions may offer a novel therapeutic avenue.

    Metabolites and their related biochemical pathways have emerged as important contributors to the development of OSA.16 An increased prevalence of OSA has been reported in patients with neurodegenerative disorders, potentially related to impairments in central respiratory control and functional alterations of the upper airway.17 The nervous system and gastrointestinal tract communicate through a bidirectional network of signaling pathways, collectively referred to as the microbiota–gut–brain axis, which involves the vagus nerve, immune mechanisms, and microbiota-associated metabolic and molecular products. Emerging evidence suggests that this axis plays a pivotal role in the pathogenesis of neurodegenerative diseases.18 These findings highlight the potential importance of microbiota–gut–brain axis related pathways in the pathogenesis and progression of OSA, suggesting that further research in this area is warranted. Previous studies have reported the involvement of metabolites and microbiota-related metabolites in the development and progression of OSA. Xu et al reported correlations between alterations in the oral microbiome and disruptions in urinary metabolites in children with OSA.19 Using Mendelian randomization (MR), Yan et al identified several gut microbiota and microbiota-related metabolites as potential independent risk factors for OSA.20 Therefore, investigating the relationship between the gut microbiota and OSA, and further exploring the potential mediating role of metabolites, is of substantial significance.

    MR provides a powerful framework for investigating the causal relationship between gut microbiota and OSA.21 In MR analysis, genetic variants are used as instrumental variables (IVs) for exposure traits, enabling the estimation of causal effects on clinical outcomes while minimizing confounding and reverse causation. In this study, we performed two-sample and two-step MR analyses using summary-level data from the genome-wide association studies (GWAS) on gut microbiota, plasma metabolites, and OSA. This study primarily focused on identifying gut microbiota and potential mediator metabolites that may confer protective effects against OSA, with the aim of uncovering novel therapeutic targets and providing preliminary insights into the underlying mechanisms.

    Methods

    Study Design

    This study was conducted using a two-sample MR framework, with single nucleotide polymorphisms (SNPs) selected as IVs. MR relies on three key assumptions: i the IVs are strongly associated with the exposure; ii the IVs are not associated with any confounders; and iii the IVs affect the outcome solely through the exposure, without exerting a direct effect on the outcome.

    An overview of the study design is provided in Figure 1. First, a two-sample MR analysis was performed to explore the potential causal relationship between gut microbiota and OSA. To reduce potential functional heterogeneity, microbial taxa within the same genus or family that showed divergent associations with the OSA were excluded. Only microbial taxa associated with a reduced risk of OSA were retained for further analysis. Second, potential mediators were identified according to the following steps. Step 1: An exploratory two-sample MR analysis was conducted to identify metabolites potentially involved in the development and progression of OSA. To account for multiple testing, the false discovery rate (FDR) was controlled using the Benjamini–Hochberg procedure. Metabolites were prioritized based on inverse variance weighted (IVW) P values adjusted for FDR (PFDR). The biological relevance of these metabolites was further assessed using the Human Metabolome Database (HMDB) (Version 5.0). Ultimately, the top 10 metabolites with potential biological significance were selected as candidate mediators for subsequent analyses (Supplementary Table 1). Step 2: Potential mediators of the association between gut microbiota and OSA were identified based on the following criteria: i a two-sample MR analysis was conducted to assess the causal relationship between each selected protective taxon and candidate mediator. P values were adjusted for multiple comparisons using the FDR correction. The directionality of the associations was confirmed to be unidirectional. ii a two-sample MR analysis was performed to evaluate the causal effect of each candidate mediator on the OSA, using a more stringent SNP selection threshold (P < 5e-6) and excluding SNPs associated with known confounders. iii the direction of the association between the mediator and OSA had to be opposite to that between the selected protective taxa and the mediator. Third, the mediating effects of the identified metabolites on the causal pathway linking gut microbiota to OSA were quantified using a two-step MR approach. In the initial step, we employed a two-sample MR analysis to assess the total direct effect of gut microbiota on OSA risk (β0). In the subsequent step, we estimated the effect of gut microbiota on plasma metabolites (β1) and the effect of plasma metabolites on OSA (β2), allowing us to calculate the indirect effect (β1 × β2). The proportion of the mediated effect was determined by dividing β1 × β2 by the total effect (β0).

    Figure 1 Overview of the study design. (A) Protective gut microbiota associated with reduced OSA risk were identified using two-sample MR analysis. (B) The top 10 candidate metabolites were selected based on FDR-adjusted IVW results and biological relevance derived from the HMDB. (C) Potential mediators were identified through a three-step process involving causal links between gut microbiota and metabolites, metabolites-OSA associations, and consistent directional effects. (D) A two-step MR analysis was conducted to quantify the mediation effect linking gut microbiota to OSA through the selected metabolites. MR analyses were primarily conducted using the IVW method, with complementary approaches including MR-Egger, weighted median, simple mode, and weighted mode. Sensitivity analyses included Cochran’s Q statistic, MR-Egger intercept test, MR-Pleiotropy Residual Sum and Outlier method, MR Steiger directionality test, and leave-one-out approach.

    Abbreviations: OSA, obstructive sleep apnea; GWAS, genome-wide association study; SNPs, single nucleotide polymorphisms; MR, mendelian randomization; IVW, inverse variance weighted; FDR, false discovery rate; PFDR, P values adjusted for FDR; HMDB, Human Metabolome Database.

    Data Sources

    We garnered the GWAS data for the gut microbiota from the Dutch Microbiome Project.22 The Dutch Microbiome Project analyzed feces from 7,738 individuals of European descent, involving 207 taxa.22 The GWAS data for plasma metabolites were obtained from the Canadian Longitudinal Study on Aging cohort, comprising 8,299 participants.23 This analysis encompassed 1,091 metabolic features and 309 metabolite ratios.

    The GWAS data for OSA were obtained from the FinnGen database (R10) with 410385 individuals of European ancestry. The data contained 43901 OSA cases and 366484 controls. OSA diagnostic criteria was depended on ICD codes (ICD-10: G47.3; ICD-9: 3472), which were obtained from the Finnish National Hospital Discharge Registry and the Causes of Death Registry. The ICD-10 code G47.3 encompasses both OSA and central sleep apnea (CSA). Nevertheless, Strausz et al validated the registry-based OSA diagnosis, reporting a positive predictive value of >98%, indicating a high level of diagnostic accuracy.24 The FinnGen study is a large-scale genomics project that has analyzed more than 500,000 Finnish biobank samples, linking genetic variations with health data to uncover disease mechanisms and predispositions.25

    Instrumental Variables Selection

    In this study, SNPs were selected as IVs, and their selection and validation followed the criteria outlined below. Firstly, due to the limited number of genome-wide significant SNPs available for gut microbiota, we applied a significance threshold of P < 1e-5 to identify potentially relevant SNPs. For the selected mediator metabolites, MR analyses were performed using OSA as the outcome, with SNPs selected based on a stricter threshold of P < 5e-6. Secondly, to minimize bias from linkage disequilibrium (LD), we performed SNP clumping using an LD threshold of R² < 0.001 within a 10,000 kb window. Thirdly, to reduce the risk of weak instrument bias, we calculated the F-statistic for each SNP and retained only those with F > 10 as valid IVs. Functional annotation of SNPs was conducted using functional mapping and gene annotation (FUMA) (Version 1.8.0), based on the most recent release of the GWAS Catalog.26 To reduce the risk of potential pleiotropy, SNPs previously associated with OSA-related traits, such as body mass index, waist circumference, hip circumference, or waist-to-hip ratio, were excluded (Supplementary Table 2).

    Statistical Analysis

    We conducted a two-sample MR analysis to evaluate the causal relationships between gut microbiota and OSA, as well as between plasma metabolites and OSA independently. We evaluated the causal association between gut microbiota and the plasma metabolites using bidirectional MR. We used the IVW method as the primary MR approach. To enhance the robustness and reliability of our findings, we also applied complementary MR methods, including MR-Egger regression, weighted median, simple mode, and weighted mode. Apart from the MR methods outlined above, we also carried out various supplementary sensitivity analyses. Firstly, we evaluated heterogeneity in causal inference through the calculation of Cochran’s Q statistic. Secondly, we applied the MR-Egger intercept test to detect horizontal pleiotropy. Moreover, the MR-Pleiotropy Residual Sum and Outlier (MR-PRESSO) method was employed to detect horizontal pleiotropy and identify potential outlier SNPs. A global test P-value < 0.05 was considered indicative of significant distortion in the causal estimates due to pleiotropy. Additionally, we employed the MR Steiger directionality test to ascertain the causal direction between the exposure and the outcome. Furthermore, the reliability of the findings was assessed through a leave-one-out approach for validation.

    Effect sizes were expressed as odds ratios (OR), β-coefficients, and corresponding 95% confidence intervals (CI). All statistical analyses were conducted using the “TwoSampleMR” package (version 0.6.6), “MRPRESSO” package, and “ggplot2” packages within R software (version 4.4.1).

    Results

    Instrument Variables Included in Analysis

    Based on the predefined selection criteria, we identified valid IVs from GWAS summary statistics of gut microbiota and plasma metabolites. Supplementary Tables 3 and 4 provide detailed characteristics of these IVs, including their corresponding F statistics. All included SNPs had F statistics greater than 10, supporting adequate instrument strength.

    Effects of Gut Microbiota on Obstructive Sleep Apnea

    Using the IVW method, this study identified 4 microbial taxa that were associated with a reduced risk of the OSA, including species Parabacteroides merdae, genus Faecalibacterium, species Faecalibacterium prausnitzii and species Bifidobacterium longum (Table 1). Species Parabacteroides merdae exhibited the most pronounced protective effect (OR = 0.909, 95% CI = 0.828~0.999, P value = 0.047). Species Faecalibacterium prausnitzii and genus Faecalibacterium showed the second strongest protective effects (species Faecalibacterium prausnitzii, OR = 0.921, 95% CI = 0.860~0.986, P value = 0.019; genus Faecalibacterium, OR = 0.923, 95% CI = 0.857~0.994, P value = 0.034). The protective effect of species Bifidobacterium longum was comparatively weaker (OR = 0.930, 95% CI = 0.869~0.997, P value = 0.040).

    Table 1 Effects of Gut Microbiota on the Risk of Obstructive Sleep Apnea

    Effects of Gut Microbiota on Mediators

    Following the established criteria and selection workflow, we identified two candidate mediators, 2-hydroxypalmitate and hyocholate, from the top 10 biologically relevant metabolites. As shown in Table 2 and Supplementary Table 5, species Parabacteroides merdae was positively associated with the levels of 2-hydroxypalmitate (β = 0.254, 95% CI = 0.117~0.391, IVW P value < 0.001, IVW PFDR value = 0.003). Since MR analyses were performed between the selected microbial taxa and the top 10 metabolites with potential biological significance, FDR correction was applied across these 10 tests using the Benjamini–Hochberg method. After FDR correction, no statistically significant associations were observed between genus Faecalibacterium and 2-hydroxypalmitate, species Faecalibacterium prausnitzii and 2-hydroxypalmitate, or species Bifidobacterium longum and hyocholate.

    Table 2 Effects of Gut Microbiota on Mediators

    Effects of Selected Mediators on the OSA and Mediation Analysis

    Table 3 and Supplementary Table 5, demonstrates that elevated levels of 2-hydroxypalmitate were associated with a reduced risk of OSA (OR = 0.926, 95% CI = 0.865~0.991, P value = 0.027). As presented in Figure 2, through two-step MR analysis, 2-hydroxypalmitate was found to mediate 20.53% of the association between the species Parabacteroides merdae and OSA.

    Table 3 Effects of Mediators on Obstructive Sleep Apnea

    Figure 2 The 2-hydroxypalmitate mediated the causal effect of species Parabacteroides merdae on OSA. Two-step MR was used to evaluate the mediating role of 2-hydroxypalmitate in the causal pathway linking species Parabacteroides merdae to OSA. The causal effect of species Parabacteroides merdae on 2-hydroxypalmitate, 2-hydroxypalmitate on OSA and species Parabacteroides merdae on OSA were assumed to be β1, β2 and β0, respectively. The proportion of the mediated effect (bold text) was determined by dividing β1× β2 by the total effect (β0). MR estimates were derived from the IVW method in two-sample MR.

    Abbreviations: OSA, obstructive sleep apnea; MR, mendelian randomization; IVW, inverse variance weighted.

    MR Sensitivity Analyses

    According to the results presented in Supplementary Table 6, no evidence of heterogeneity was observed, as indicated by Cochran’s Q statistic (all P values > 0.05). Both the MR-Egger intercept test and the MR-PRESSO global test indicated no signs of horizontal pleiotropy, with P values greater than 0.05. In addition, the MR-PRESSO method did not identify any outlier variants. The MR-Steiger directionality test provided no evidence supporting a reverse causal relationship from the OSA to the four protective taxa or to 2-hydroxypalmitate. As shown in Supplementary Figures 13, the leave-one-out analysis suggested that certain SNPs may have disproportionately influenced the causal estimates. Based on the overall findings from our sensitivity analyses, the observed associations appear to be relatively robust. Nonetheless, these results should be interpreted with caution.

    Discussion

    In this study, we performed a comprehensive MR analysis utilizing large-scale GWAS summary data to explore the causal relationships between gut microbiota, plasma metabolites, and OSA. In the Dutch Microbiome Project, species Parabacteroides merdae, genus Faecalibacterium, species Faecalibacterium prausnitzii and species Bifidobacterium longum demonstrated a potential protective association with OSA. This study was exploratory in nature. We included the top 10 metabolites with potential biological significance as candidate mediators. Among them, only 2-hydroxypalmitate was found to mediate the association between species Parabacteroides merdae and OSA, with a mediation proportion of 20.53%.

    Species Parabacteroides Merdae and OSA

    Species Parabacteroides merdae is a Gram-negative, anaerobic, rod-shaped bacterium commonly found in the human gut microbiota.27 Consistent with our findings, species Parabacteroides merdae retained a strong negative association with OSA.28 The potential protective effect of species Parabacteroides merdae against OSA may be related to its role in alleviating obesity.29 Qiao et al reported that species Parabacteroides merdae significantly attenuated high-fat diet-induced weight gain in mice.30 Given that obesity is a major risk factor for OSA, the beneficial effect of species Parabacteroides merdae on obesity may partially explain its protective role against OSA. Fecal microbiota transplantation has been shown to lower systolic blood pressure (SBP), with increased abundances of species Parabacteroides merdae associated with SBP reduction.31 Given that OSA is a known risk factor for hypertension, it is plausible that the SBP-lowering effect of species Parabacteroides merdae may be related to improvements in nocturnal intermittent hypoxia. However, this hypothesis requires further investigation. Evidence linking species Parabacteroides merdae to OSA remains limited. However, further investigation into its potential protective role may help uncover novel therapeutic strategies. Given the complexity of the underlying mechanisms, further research is warranted to identify the key pathways and molecular targets through which species Parabacteroides merdae may influence OSA progression.

    2-Hydroxypalmitate and OSA

    2-hydroxypalmitate, also known as 2-hydroxyhexadecanoic acid, has emerged as a representative member of the 2-hydroxy fatty acid family.32 These molecules constitute essential structural components of mammalian sphingolipids. Sphingolipids are essential components of cell membranes, particularly abundant in the brain, where they help maintain myelin integrity, facilitate neuronal signaling, and support intercellular communication.33 Fatty acid 2-hydroxylase (FA2H) catalyzes the biosynthesis of hydroxylated sphingolipids by introducing a hydroxyl group at the α-carbon of long-chain fatty acids.34 FA2H gene mutations are linked to leukodystrophy and spastic paraparesis, highlighting the importance of hydroxylated fatty acid-containing sphingolipids in nervous system function.35,36 Increasing evidence suggests that alterations in sphingolipid pathways may contribute to the etiopathogenesis of neurodegenerative diseases.37 Parkinson’s disease (PD) is one of the most prevalent neurodegenerative disorders. Sphingolipids play a critical role in various cellular processes involved in the pathogenesis of PD, including mitochondrial function, autophagy, and endosomal trafficking.38 Upper airway obstruction and dysfunction are observed in approximately 24% to 65% of PD patients, primarily attributed to laryngopharyngeal motor impairment.39,40 This dysfunction reflects a broader impairment of neuromuscular control in the upper airway, characterized by reduced tone in the pharyngeal dilator muscles, impaired coordination of respiratory and swallowing reflexes, and delayed glottic opening.17 These abnormalities may predispose individuals with PD to the development of OSA, particularly during sleep when compensatory mechanisms are further diminished. Given the essential role of sphingolipids in maintaining neuronal membrane stability and supporting axonal conduction, their dysregulation may plausibly impair the integrity of central or peripheral neural pathways involved in upper airway motor control. However, it must be acknowledged that current evidence directly linking 2-hydroxypalmitate to the regulation of upper airway motor function is limited. Further studies are warranted to clarify this potential association.

    However, some studies have suggested that 2-hydroxypalmitate may contribute to the development of OSA. 2-hydroxypalmitate is associated with dyslipidemia, a condition commonly observed in obesity.41,42 Given that obesity is a major risk factor for OSA, 2-hydroxypalmitate may contribute to the development of OSA through pathways related to fatty acid metabolism. Sullivan et al43 found that vitamin D supplementation was associated with lower 2-hydroxypalmitate levels, while Ayyıldız et al44 reported a potential beneficial effect of vitamin D supplementation on the prognosis of mild OSA. These findings are inconsistent with our results. Nonetheless, given the exploratory nature of this study, further research is needed to clarify the role of 2-hydroxypalmitate in OSA pathogenesis.

    2-Hydroxypalmitate as a Putative Mediator: Implications and Future Directions

    In this study, we identified 2-hydroxypalmitate as a potential mediator of the relationship between species Parabacteroides merdae and the reduced risk of OSA. The mediation analysis demonstrated that 2-hydroxypalmitate accounted for 20.53% of the total effect. In terms of effect size, this proportion represents a moderate mediation effect, comparable to those observed in other microbial-metabolite axes involved in disease progression. For example, prior studies have reported mediation proportions ranging from 6.5% to 25.1% in obesity,45 10.29% to 21.9% in type 2 diabetes,46 11.04% to 15.35% in PD,47 14.62% to 37.48% in chronic airway disease,48,49 and 8.1% to 22.8% in various cancer types.50 These findings suggest that the mediation effect observed in our study falls within a biologically meaningful range and supports the role of 2-hydroxypalmitate as a mediator within the causal pathway from species Parabacteroides merdae to OSA.

    Previous studies suggest that species Parabacteroides merdae may reduce the risk of OSA through anti-obesity effects, while 2-hydroxypalmitate may confer protection via sphingolipid metabolism, which modulates upper airway motor function. In our analysis, species Parabacteroides merdae and 2-hydroxypalmitate were positively correlated and both linked to reduced OSA risk, supporting the plausibility of this mediation pathway. Although direct evidence for the regulatory relationship between species Parabacteroides merdae and 2-hydroxypalmitate is currently lacking, our findings suggest a biologically relevant connection that merits further investigation. Overall, this study reveals a gut microbiota-metabolite-OSA pathway and offers new evidence for microbial metabolic mediation in OSA. Given the exploratory nature of this study, future experimental and longitudinal studies are warranted to validate these results and elucidate the underlying mechanisms.

    Limitations

    This study has several limitations that should be acknowledged. First, the GWAS summary statistics used in our MR analyses were derived exclusively from individuals of European ancestry. As a result, our findings may not be fully generalizable to other populations, given known differences in genetic architecture, LD structure, allele frequencies, and environmental modifiers across ancestries. These population-specific factors could influence both the strength and direction of causal associations, potentially limiting the applicability of our results to non-European groups. To enhance the external validity of microbiota-metabolite-disease mediation frameworks, future studies should incorporate GWAS data from ancestrally diverse cohorts and perform replication analyses across multiple populations. Such efforts are essential to improve the global relevance and translational potential of microbiome-informed causal inference. Second, sleep apnea in the FinnGen study was defined using ICD codes (ICD-10: G47.3; ICD-9: 3472), and individual-level polysomnographic (PSG) data, such as the apnea-hypopnea index, oxygen desaturation index, and sleep architecture, were not available. This limitation precludes accurate confirmation of OSA diagnoses, assessment of disease severity, and identification of clinical subtypes (eg, rapid eye movement-predominant or positional OSA). Notably, ICD-10 code G47.3 includes both OSA and CSA. While Strausz et al24 validated the registry-based OSA definition and reported high diagnostic accuracy, the potential for misclassification remains, particularly due to the inclusion of CSA cases. However, given the low prevalence of CSA51 and the lack of evidence linking our exposures of interest to CSA risk, any such misclassification is likely nondifferential with respect to exposure. From a statistical perspective, this would tend to bias the effect estimates toward the null, resulting in a conservative estimate of the true causal effect. Future studies incorporating PSG-confirmed, individual-level OSA data are therefore warranted to validate and refine these findings. Third, a relaxed significance threshold (P < 1e-5) was used for SNP selection in the MR analyses to increase the number of valid IVs and improve statistical power. This approach has been adopted in previous MR studies involving complex traits and microbiota-related exposures,50,52 and is considered acceptable in exploratory settings according to a strategy supported by previous methodological literature.53–55 However, the use of a relaxed threshold may increase the risk of including weak or pleiotropic IVs. To mitigate this concern, we retained only SNPs with F > 10 to ensure sufficient IVs strength, and excluded those associated with known confounders whenever possible. In addition, the leave-one-out analysis suggested that one or more SNPs might disproportionately influence the causal estimates. Nonetheless, no evidence of heterogeneity (Cochran’s Q statistic) or horizontal pleiotropy (MR-Egger intercept and MR-PRESSO global test) was observed, supporting the robustness of our results. These findings should be interpreted with caution. Future studies using larger GWAS datasets and more advanced MR methods are needed to validate our conclusions. For example, multivariable MR, summary-data-based MR to explore the relationships among genetic variants, exposures, and outcomes,56,57 and Bayesian co-localization analysis to verify shared causal variants and strengthen causal inference58 may offer further insights.

    Conclusions

    In summary, this study highlights the protective effect of species Parabacteroides merdae against OSA. 2-hydroxypalmitate may act as a partial mediator in the association between species Parabacteroides merdae and OSA. These findings provide novel insights into the mechanisms underlying OSA and suggest potential therapeutic targets, offering promising directions for future research and clinical interventions.

    Abbreviations

    OSA, Obstructive sleep apnea; MR, Mendelian randomization; IVs, instrumental variables; GWAS, genome-wide association studies; SNPs, single-nucleotide polymorphisms; FDR, false discovery rate; IVW, inverse variance weighted; PFDR, P values adjusted for false discovery rate; HMDB, Human Metabolome Database; CSA, central sleep apnea; LD, linkage disequilibrium; FUMA, functional mapping and gene annotation; MR-PRESSO, the MR-Pleiotropy Residual Sum and Outlier; OR, odds ratios; CI, confidence intervals; SBP, systolic blood pressure; FA2H, Fatty acid 2-hydroxylase; PD, Parkinson’s disease; PSG, polysomnographic.

    Data Sharing Statement

    The datasets supporting the conclusions of this article are available in IEU open GWAS project repository (https://www.ebi.ac.uk/gwas/). Details regarding the GWAS on OSA can be accessed through the following link: https://r10.risteys.finngen.fi/endpoints/G6_SLEEPAPNO/.

    Ethics Approval and Consent to Participate

    All data in this study were derived from publicly accessible GWAS using anonymized summary-level datasets. As this research exclusively analyzed de-identified aggregate data without involving individual participants, sensitive personal information, or commercial interests, ethical approval was formally waived by the Ethics Committee of Beijing Friendship Hospital, Capital Medical University (Approval No.: 2025-P2-115). This study was conducted in full compliance with the Declaration of Helsinki and relevant national ethical regulations.

    Acknowledgments

    We extend our sincere gratitude to the authors and participants of all GWAS studies whose summary statistics data were utilized in this research.

    Author Contributions

    Xiaona Wang: Conceptualization, Data Curation, Formal analysis, Funding acquisition, Investigation, Methodology, Software, Validation, and Writing – original draft. Ranran Zhao, Jia Guo, and Ke Yang: Data Curation, Formal analysis, Investigation, Methodology, Validation, and Writing – review & editing. Bo Xu: Conceptualization, Funding acquisition, Methodology, Project administration, Resources, Software, Supervision, and Writing – review & editing. All authors made significant contributions to the study, approved the final manuscript for submission, agreed on the choice of journal, and take full responsibility for the integrity of the work.

    Funding

    This work was supported by the Capital’s Funds for Health Improvement and Research (2024-2-1101 20240402105353, China) and the Seed Program of Beijing Friendship Hospital (YYZZ202235).

    Disclosure

    The authors declare that they have no competing interests.

    References

    1. Heinzer R, Vat S, Marques-Vidal P, et al. Prevalence of sleep-disordered breathing in the general population: the HypnoLaus study. Lancet Respir Med. 2015;3(4):310–318. doi:10.1016/s2213-2600(15)00043-0

    2. Neelapu BC, Kharbanda OP, Sardana HK, et al. Craniofacial and upper airway morphology in adult obstructive sleep apnea patients: a systematic review and meta-analysis of cephalometric studies. Sleep Med Rev. 2017;31:79–90. doi:10.1016/j.smrv.2016.01.007

    3. White DP. Pathogenesis of obstructive and central sleep apnea. Am J Respir Crit Care Med. 2005;172(11):1363–1370. doi:10.1164/rccm.200412-1631SO

    4. Lv R, Liu X, Zhang Y, et al. Pathophysiological mechanisms and therapeutic approaches in obstructive sleep apnea syndrome. Signal Transduct Target Ther. 2023;8(1):218. doi:10.1038/s41392-023-01496-3

    5. Cai Y, Juszczak HM, Cope EK, Goldberg AN. The microbiome in obstructive sleep apnea. Sleep. 2021;44(8). doi:10.1093/sleep/zsab061

    6. Silva V, Palacios-Muñoz A, Okray Z, et al. The impact of the gut microbiome on memory and sleep in Drosophila. J Exp Biol. 2021;224(Pt 3). doi:10.1242/jeb.233619

    7. Grosicki GJ, Riemann BL, Flatt AA, Valentino T, Lustgarten MS. Self-reported sleep quality is associated with gut microbiome composition in young, healthy individuals: a pilot study. Sleep Med. 2020;73:76–81. doi:10.1016/j.sleep.2020.04.013

    8. Fei N, Choo-Kang C, Reutrakul S, et al. Gut microbiota alterations in response to sleep length among African-origin adults. PLoS One. 2021;16(9):e0255323. doi:10.1371/journal.pone.0255323

    9. O’Connor KM, Lucking EF, Golubeva AV, et al. Manipulation of gut microbiota blunts the ventilatory response to hypercapnia in adult rats. EBioMedicine. 2019;44:618–638. doi:10.1016/j.ebiom.2019.03.029

    10. Ridaura VK, Faith JJ, Rey FE, et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science. 2013;341(6150):1241214. doi:10.1126/science.1241214

    11. Andersen ML, Gozal D, Pires GN, Tufik S. Exploring the potential relationships among obstructive sleep apnea, erectile dysfunction, and gut microbiota: a narrative review. Sex Med Rev. 2023;12(1):76–86. doi:10.1093/sxmrev/qead026

    12. Badran M, Khalyfa A, Ericsson AC, et al. Gut microbiota mediate vascular dysfunction in a murine model of sleep apnoea: effect of probiotics. Eur Respir J. 2023;61(1):2200002. doi:10.1183/13993003.00002-2022

    13. Liu Q, Liu Y, Li F, et al. Probiotic culture supernatant improves metabolic function through FGF21-adiponectin pathway in mice. J Nutr Biochem. 2020;75:108256. doi:10.1016/j.jnutbio.2019.108256

    14. Liu J, Li T, Wu H, et al. Lactobacillus rhamnosus GG strain mitigated the development of obstructive sleep apnea-induced hypertension in a high salt diet via regulating TMAO level and CD4(+) T cell induced-type I inflammation. Biomed Pharmacother. 2019;112:108580. doi:10.1016/j.biopha.2019.01.041

    15. Xu H, Wang J, Cai J, et al. Protective Effect of Lactobacillus rhamnosus GG and its Supernatant against Myocardial Dysfunction in Obese Mice Exposed to Intermittent Hypoxia is Associated with the Activation of Nrf2 Pathway. Int J Biol Sci. 2019;15(11):2471–2483. doi:10.7150/ijbs.36465

    16. Tomar MS, Araniti F, Sahai PK, et al. Urinary metabolite signatures reflect the altered host metabolism in severe obstructive sleep apnea. J Chromatogr B Analyt Technol Biomed Life Sci. 2023;1231:123938. doi:10.1016/j.jchromb.2023.123938

    17. Bahia C, Pereira JS. Obstructive sleep apnea and neurodegenerative diseases: a bidirectional relation. Dement Neuropsychol. 2015;9(1):9–15. doi:10.1590/s1980-57642015dn91000003

    18. Loh JS, Mak WQ, Tan LKS, et al. Microbiota-gut-brain axis and its therapeutic applications in neurodegenerative diseases. Signal Transduct Target Ther. 2024;9(1):37. doi:10.1038/s41392-024-01743-1

    19. Xu H, Li X, Zheng X, et al. Pediatric Obstructive Sleep Apnea is Associated With Changes in the Oral Microbiome and Urinary Metabolomics Profile: a Pilot Study. J Clin Sleep Med. 2018;14(9):1559–1567. doi:10.5664/jcsm.7336

    20. Yan W, Jiang M, Hu W, et al. Causality Investigation between Gut Microbiota, Derived Metabolites, and Obstructive Sleep Apnea: a Bidirectional Mendelian Randomization Study. Nutrients. 2023;15(21):4544. doi:10.3390/nu15214544

    21. Emdin CA, Khera AV, Kathiresan S. Mendelian Randomization. JAMA. 2017;318(19):1925–1926. doi:10.1001/jama.2017.17219

    22. Lopera-Maya EA, Kurilshikov A, van der Graaf A, et al. Effect of host genetics on the gut microbiome in 7,738 participants of the Dutch Microbiome Project. Nat Genet. 2022;54(2):143–151. doi:10.1038/s41588-021-00992-y

    23. Chen Y, Lu T, Pettersson-Kymmer U, et al. Genomic atlas of the plasma metabolome prioritizes metabolites implicated in human diseases. Nat Genet. 2023;55(1):44–53. doi:10.1038/s41588-022-01270-1

    24. Strausz S, Ruotsalainen S, Ollila HM, et al. Genetic analysis of obstructive sleep apnoea discovers a strong association with cardiometabolic health. Eur Respir J. 2021;57(5):2003091. doi:10.1183/13993003.03091-2020

    25. Kurki MI, Karjalainen J, Palta P, et al. FinnGen provides genetic insights from a well-phenotyped isolated population. Nature. 2023;613(7944):508–518. doi:10.1038/s41586-022-05473-8

    26. Watanabe K, Taskesen E, van Bochoven A, Posthuma D. Functional mapping and annotation of genetic associations with FUMA. Nat Commun. 2017;8(1):1826. doi:10.1038/s41467-017-01261-5

    27. Sakamoto M, Benno Y. Reclassification of Bacteroides distasonis, Bacteroides goldsteinii and Bacteroides merdae as Parabacteroides distasonis gen. nov. comb. nov. Parabacteroides goldsteinii comb. nov. and Parabacteroides merdae comb. nov. Int J Syst Evol Microbiol. 2006;56(Pt 7):1599–1605. doi:10.1099/ijs.0.64192-0

    28. Liu L, He G, Yu R, et al. Causal relationships between gut microbiome and obstructive sleep apnea: a bi-directional Mendelian randomization. Front Microbiol. 2024;15:1410624. doi:10.3389/fmicb.2024.1410624

    29. Sugawara Y, Kanazawa A, Aida M, Yoshida Y, Yamashiro Y, Watada H. Association of gut microbiota and inflammatory markers in obese patients with type 2 diabetes mellitus: post hoc analysis of a synbiotic interventional study. Biosci Microbiota Food Health. 2022;41(3):103–111. doi:10.12938/bmfh.2021-081

    30. Qiao S, Liu C, Sun L, et al. Gut Parabacteroides merdae protects against cardiovascular damage by enhancing branched-chain amino acid catabolism. Nat Metab. 2022;4(10):1271–1286. doi:10.1038/s42255-022-00649-y

    31. Fan L, Chen J, Zhang Q, et al. Fecal microbiota transplantation for hypertension: an exploratory, multicenter, randomized, blinded, placebo-controlled trial. Microbiome. 2025;13(1):133. doi:10.1186/s40168-025-02118-6

    32. Hama H. Fatty acid 2-Hydroxylation in mammalian sphingolipid biology. Biochim Biophys Acta. 2010;1801(4):405–414. doi:10.1016/j.bbalip.2009.12.004

    33. van Echten-Deckert G, Herget T. Sphingolipid metabolism in neural cells. Biochim Biophys Acta. 2006;1758(12):1978–1994. doi:10.1016/j.bbamem.2006.06.009

    34. Guo L, Zhang X, Zhou D, Okunade AL, Su X. Stereospecificity of fatty acid 2-hydroxylase and differential functions of 2-hydroxy fatty acid enantiomers. J Lipid Res. 2012;53(7):1327–1335. doi:10.1194/jlr.M025742

    35. Dick KJ, Eckhardt M, Paisán-Ruiz C, et al. Mutation of FA2H underlies a complicated form of hereditary spastic paraplegia (SPG35). Hum Mutat. 2010;31(4):E1251–60. doi:10.1002/humu.21205

    36. Edvardson S, Hama H, Shaag A, et al. Mutations in the fatty acid 2-hydroxylase gene are associated with leukodystrophy with spastic paraparesis and dystonia. Am J Hum Genet. 2008;83(5):643–648. doi:10.1016/j.ajhg.2008.10.010

    37. van Kruining D, Luo Q, van Echten-Deckert G, et al. Sphingolipids as prognostic biomarkers of neurodegeneration, neuroinflammation, and psychiatric diseases and their emerging role in lipidomic investigation methods. Adv Drug Deliv Rev. 2020;159:232–244. doi:10.1016/j.addr.2020.04.009

    38. Vos M, Klein C, Hicks AA. Role of ceramides and sphingolipids in Parkinson’s disease. J Mol Biol. 2023;435(12):168000. doi:10.1016/j.jmb.2023.168000

    39. Herer B, Arnulf I, Housset B. Effects of levodopa on pulmonary function in Parkinson’s disease. Chest. 2001;119(2):387–393. doi:10.1378/chest.119.2.387

    40. Yu Q, Hu X, Zheng T, et al. Obstructive sleep apnea in Parkinson’s disease: a prevalent, clinically relevant and treatable feature. Parkinsonism Relat Disord. 2023;115:105790. doi:10.1016/j.parkreldis.2023.105790

    41. Gu X, Li C, He J, et al. Serum metabolites associate with lipid phenotypes among Bogalusa Heart Study participants. Nutr Metab Cardiovasc Dis. 2020;30(5):777–787. doi:10.1016/j.numecd.2020.01.004

    42. Nussbaumerova B, Rosolova H. Obesity and Dyslipidemia. Curr Atheroscler Rep. 2023;25(12):947–955. doi:10.1007/s11883-023-01167-2

    43. Sullivan VK, Chen J, Bernard L, et al. Serum and urine metabolite correlates of vitamin D supplementation in the Atherosclerosis Risk in Communities (ARIC) study. Clin Nutr ESPEN. 2025;67:523–532. doi:10.1016/j.clnesp.2025.03.172

    44. Ayyıldız F, Yıldıran H, Afandiyeva N, Gülbahar Ö, Köktürk O. The effects of vitamin D supplemantation on prognosis in patients with mild obstructive sleep apnea syndrome. Turk J Med Sci. 2021;51(5):2524–2533. doi:10.3906/sag-2101-83

    45. Li X, Wu Q, Liu S. Identification of 1,400 metabolites as mediators of obesity in 473 gut microbiota taxa: a mediation Mendelian randomization study. Microbiol Spectr. 2025;13(6):e0189224. doi:10.1128/spectrum.01892-24

    46. Zheng X, Chen M, Zhuang Y, et al. Genetic associations between gut microbiota and type 2 diabetes mediated by plasma metabolites: a Mendelian randomization study. Front Endocrinol. 2024;15:1430675. doi:10.3389/fendo.2024.1430675

    47. Chen J, Zhu L, Wang F, et al. Plasma metabolites as mediators between gut microbiota and Parkinson’s disease: insights from Mendelian randomization. Mol Neurobiol. 2025;62(6):7945–7956. doi:10.1007/s12035-025-04765-0

    48. Du Y, Wang S, Zhou T, Zhao Z. Causal effects of gut microbiota and metabolites on chronic obstructive pulmonary disease: a bidirectional two sample Mendelian randomization study. Int J Chron Obstruct Pulmon Dis. 2024;19:2153–2167. doi:10.2147/copd.S472218

    49. Zheng X, Yin J, Zhao L, Qian Y, Xu J. Mediation analysis of gut microbiota and plasma metabolites in asthma pathogenesis using Mendelian randomization. J Asthma. 2025;62(8):1351–1363. doi:10.1080/02770903.2025.2478504

    50. Luan B, Yang Y, Yang Q, et al. Gut microbiota, blood metabolites, & pan-cancer: a bidirectional Mendelian randomization & mediation analysis. AMB Express. 2025;15(1):59. doi:10.1186/s13568-025-01866-w

    51. Roberts EG, Raphelson JR, Orr JE, LaBuzetta JN, Malhotra A. The pathogenesis of central and complex sleep apnea. Curr Neurol Neurosci Rep. 2022;22(7):405–412. doi:10.1007/s11910-022-01199-2

    52. Lv WQ, Lin X, Shen H, et al. Human gut microbiome impacts skeletal muscle mass via gut microbial synthesis of the short-chain fatty acid butyrate among healthy menopausal women. J Cachexia Sarcopenia Muscle. 2021;12(6):1860–1870. doi:10.1002/jcsm.12788

    53. Burgess S, Butterworth A, Thompson SG. Mendelian randomization analysis with multiple genetic variants using summarized data. Genet Epidemiol. 2013;37(7):658–665. doi:10.1002/gepi.21758

    54. Burgess S, Thompson SG. Mendelian Randomization: Methods for Using Genetic Variants in Causal Estimation. CRC Press; 2015.

    55. Davies NM, Holmes MV, Davey Smith G. Reading Mendelian randomisation studies: a guide, glossary, and checklist for clinicians. BMJ. 2018;362:k601. doi:10.1136/bmj.k601

    56. Zhu Z, Zhang F, Hu H, et al. Integration of summary data from GWAS and eQTL studies predicts complex trait gene targets. Nat Genet. 2016;48(5):481–487. doi:10.1038/ng.3538

    57. Zhao C, Fan Z, Zhang R, Sun Y, Li WY. The role of the major histocompatibility complex region on chromosome 6 in skin atrophy: a Mendelian randomization study. J Cosmet Dermatol. 2025;24(3):e70040. doi:10.1111/jocd.70040

    58. Mao R, Li J, Xiao W. Identification of prospective aging drug targets via Mendelian randomization analysis. Aging Cell. 2024;23(7):e14171. doi:10.1111/acel.14171

    Continue Reading

  • Prevention of eating disorders with single session interventions: Hype or promise?

    Prevention of eating disorders with single session interventions: Hype or promise?

    Is reducing the risk of eating disorders with single session interventions a hype or a promise? Tracey Wade at the Flinders University Institute for Mental Health and Wellbeing investigates

    What is a single session intervention?

    Single session interventions (SSIs) are brief, structured, goal-oriented programs, following evidence-based approaches to create meaningful change to mental health in a single encounter.

    What evidence supports SSIs?

    An umbrella review of 24 systematic reviews and meta-analyses of SSIs was published between 2007 and 2024. (1) Only seven included trials of digital or paper-based self-guided SSIs; the remainder focused on face-to-face interactions. Overall, SSIs showed a small, significant positive effect across outcomes and age groups.

    Compared to anxiety, depression and substance use, the evidence supporting SSIs for disordered eating is sparse: only two of the systematic reviews included in the umbrella review included studies evaluating eating problems, and only one reported positive effects in adolescents and adults.

    What is the rationale for use in the prevention of eating disorders?

    So far, insufficient evidence exists to support the use of unguided digital SSIs in the prevention of disordered eating. The rationale for further investigation, however, does exist, as shown in the Figure. Significantly, SSIs may successfully decrease dietary restriction, a key risk factor for the development of disordered eating. Dietary restriction refers to consciously trying to cut back the overall amount eaten to influence shape or weight.

    Two studies show a decrease in dietary restriction after completing a SSI. One showed a reduction in restrictive eating in depressed adolescents at three-month follow-up compared to a control condition when either completing a SSI on Behavioural Activation (doing activities that are considered pleasant or display some mastery) or an introduction to the brain and a lesson on neuroplasticity. (2) The second showed a decrease in dietary restriction in adults (mean age of 27.99 years) seeking treatment for an eating disorder, using adapted versions of the two SSIs from the previous study (3) (shown in figure 1).

    Table 2

    What questions need to be answered?

    A variety of questions that need to be addressed in terms of the usefulness of SSIs for the prevention of eating disorders are listed in table 1. (4,5)

    Figure 1

    Our current research

    Our research, funded by a National Mental Health and Research Council Investigator Grant (2025665), has developed nine SSIs on a smartphone app. We consulted young people and members of our Expert Advisory Group (people with lived experience, significant others, and clinicians and researchers specialising in eating disorders) to ensure that the content and features were as engaging as possible. (6)

    Young people aged 14–25 years old with elevated weight concern will be randomised to one of nine SSIs tackling risk factors for disordered eating or a control condition. The content of each SSI is described below. This important research will help better understand the role SSIs can play in reducing the risk of eating disorders in youth.

    References

    1. Schleider JL, Zapata JP, Rapoport A., et al. (2025). Single-Session Interventions for Mental Health Problems and Service Engagement: Umbrella Review of Systematic Reviews and Meta-Analyses. Annual Review of Clinical Psychology, 21(1), 279–303.
    2. Schleider JL., Mullarkey MC, Fox KR, et al. (2022). A randomized trial of online single-session interventions for adolescent depression during COVID-19. Nature Human Behaviour, 6(2), 258–268.
    3. Wade TD, Waller G. (2025). Transdiagnostic single session interventions identify rapid versus gradual responders and inform therapy personalisation before commencing therapy for eating disorders. Cognitive Behaviour Therapy.
    4. O’Dea B & de Valle MK. (2025). Trial, Error, and Insight: Using the Pilot Study of the HOPE Program to Inform Next Steps for Digital Single-Session Research for Eating Disorders. International Journal of Eating Disorders, 10.1002/eat.24487.
    5. Thompson M, Radunz M, Wade TD, Balzan RP. (2024). Bridging the gap: Can single session interventions help enhance mental health treatment delivery for young people in Australia? Australian & New Zealand Journal of Psychiatry, 58(10):829-830.
    6. Pellizzer ML, Pennesi J-L, Radunz M, Zhou Y, Wade TD. (2025). Piloting single session interventions in a sample of weight-concerned youth: Study protocol for a randomised controlled trial. Body Image, 54, 101945.

    Continue Reading

  • Showcasing innovation in health, mining and urban well-being

    Showcasing innovation in health, mining and urban well-being

    image: ©imaginima | iStock

    The sixth edition of the European Research and Innovation (R&I) Days will take place on 16-17 September 2025 at The Square in Brussels, bringing together key players from across Europe’s research, business, and innovation

    Organised by the European Commission, this event will help to shape the future of EU research by fostering dialogue, collaboration, and solutions in areas critical to Europe’s competitiveness and sustainability.

    Among the highlights of the 2025 edition will be three groundbreaking projects managed by the European Health and Digital Executive Agency (HaDEA). These projects, funded under the Horizon 2020 programme, focus on advancing research in health, digital innovation, and environmental sustainability, key priorities for the EU’s future.

    MAESTRIA: Advancing heart health through AI

    A significant project featured at the event is MAESTRIA (Machine Learning Artificial Intelligence Early Detection of Stroke and Atrial Fibrillation). This project is transforming how medical professionals detect and manage atrial cardiomyopathy, a heart condition associated with atrial fibrillation (AF) and stroke.

    By combining advanced AI algorithms, medical imaging, and cutting-edge research, MAESTRIA is building the first-ever digital diagnostic platform specifically for atrial cardiomyopathy. The goal is to enable personalised diagnosis, better risk assessment, and more effective treatment options, ultimately reducing complications like AF and stroke.

    The project represents a significant leap forward in preventive healthcare, offering scalable tools to enhance patient outcomes and alleviate the burden of cardiovascular disease across Europe.

    PERSEPHONE: Robotics for sustainable seep mining

    In terms of industrial innovation, PERSEPHONE (Autonomous Exploration and Extraction of Deep Mineral Deposits) is leading efforts to modernise and automate deep mining operations. As demand for raw materials critical to the green and digital transitions grows, Europe is turning to more brilliant, safer, and more sustainable ways to access these resources.

    PERSEPHONE is developing compact, energy-efficient autonomous robots capable of exploring deep or abandoned mines. These robotic systems are equipped with sensors and cameras that generate 3D scans of underground environments. The data collected is used to create a digital twin of the mine, enabling precise planning for safe and efficient mineral extraction.

    By minimising risks to human workers and reducing environmental impact, PERSEPHONE supports the EU’s efforts to build a resilient supply chain for critical raw materials, essential for clean energy technologies and digital innovation.

    eMOTIONAL cities: Designing healthier urban spaces

    Another innovative project, eMOTIONAL Cities, recently concluded its mission to understand how urban environments affect people’s emotional and mental well-being. Using a unique device known as the Cities walker backpack, the project gathered real-life biometric data to assess how individuals respond emotionally to different urban settings.

    The findings from eMOTIONAL Cities offer valuable insights for urban planners and policymakers aiming to create healthier, more inclusive cities. By linking emotional responses to environmental factors, the project contributes to the design of urban spaces that promote calmness, happiness, and social connection.

    R&I days 2025

    Attendees of R&I Days 2025 will have the chance to explore these projects in the exhibition area, engage in high-level discussions, and participate in networking opportunities with policymakers, researchers, and innovators.

    This year’s event highlights how EU-funded research, under programmes such as Horizon 2020 and Horizon Europe, continues to drive forward solutions in health, digitalisation, and sustainability, paving the way for a stronger, more resilient Europe.

    Continue Reading

  • ACS report shows prostate cancer remains a significant public health challenge

    ACS report shows prostate cancer remains a significant public health challenge

    The American Cancer Society (ACS) has published its annual Prostate Cancer Statistics, 2025 report, showing increasing incidence rates alongside slowing declines in mortality rates.1

    Data for the report were collected by the National Cancer Institute and the Centers for Disease Control and Prevention. Population-based incidence data were analyzed through 2021, and mortality data were analyzed through 2023.

    The ACS estimates that in 2025, there will be 313,780 new cases of prostate cancer and 35,770 deaths due to the disease.

    Trends in Incidence and Mortality

    Overall, the data showed a marked reversal in prostate cancer incidence trends in recent years. Although the incidence rates declined by 6.4% per year from 2007 to 2014, data show that these rates have been increasing by 3.0% annually from 2014 to 2021. This trend has been largely driven by increasing rates of advanced-stages diagnoses, which are climbing by approximately 4.6% to 4.8% per year.

    Notably, prostate cancer mortality rates have been decreasing since the 1990s, though the declines have slowed in recent years. In the 1990s and 2000s, mortality rates decreased by 3% to 4% annually. Over the past decade, these rates have waned to approximately 0.6% per year.

    According to the report, rates of distant-stage disease are increasing across every age group. Specifically, rates are increasing by about 3% in those younger than 55 years and about 6% among those aged 55 years and older.

    The 5-year relative survival rate for distant-stage prostate cancer is 38%, but the rate increases to nearly 100% for men diagnosed with local-stage or regional-stage disease, emphasizing the importance of early detection.

    Overall, the ACS estimates that in 2025, there will be 313,780 new cases of prostate cancer and 35,770 deaths due to the disease.

    Persistent Disparities

    The report also sheds light on persistent and wide racial disparities.

    Based on the data, Black men have a 67% higher incidence rate and are 2 times more likely to die from prostate cancer compared with White men. Similarly, Native American men have a 12% higher prostate cancer mortality rate compared with White men, despite having a 13% lower incidence rate.

    “Our research highlighting the continued increases in prostate cancer incidence and persistent racial disparities underscores the need for redoubled efforts to understand the etiology of prostate cancer and optimize early detection,” commented lead author Tyler Kratzer, MPH, associate scientist, cancer surveillance research at the ACS, in a news release from the organization.2 “At age 50, per ACS guidelines, all men should have a conversation with their health care provider about the benefits and harms of screening, but Black men and those with a family history of prostate cancer should have that conversation at age 45.”

    Data also showed that American Indian and Alaska Native (AIAN) men were the most likely racial group to be diagnosed with distant-stage disease (12% vs 8% among White men). According to the authors, this finding “at least in part reflect[s] lower screening prevalence compared with other men.”

    Further, data showed that prostate cancer mortality rates are highest among Black men at 36.9 per 100,000 population, following by 20.6 deaths per 100,000 among AIAN men, 18.4 deaths per 100,000 among White men, 15.4 deaths per 100,000 among Hispanic men, and 8.8 deaths per 100,000 among Asian American and Pacific Islander men.

    By geographic location, the highest prostate cancer mortality rates were observed in Washington DC (27.5 deaths per 100,000 population) and Mississippi (24.8 deaths per 100,000 population), both of which have a high proportion of Black residents.

    The authors noted, “Increases in advanced diagnosis and persistent disparities highlight the need for redoubled efforts to optimize early detection and address barriers to equitable outcomes, including improved access to high-quality health care for all men.”

    Ongoing Legislative Efforts

    The Prostate-Specific Antigen Screening (PSA) for High-risk Insured Men (HIM) Act (H.R. 1300/S. 297) is a bipartisan bill in Congress aimed at improving access to prostate cancer screening. Specifically, this bill would waive cost-sharing requirements (deductibles, copays, and coinsurance) for prostate cancer screening tests for men who are at high-risk for the disease.

    The ACS Cancer Action Network, the advocacy affiliate for the ACS, has expressed strong support for the bill.

    “Out-of-pocket costs such as co-pays can be a barrier to accessing early detection,” explained Lisa A. Lacasse, president of ACS CAN, in the news release.2 “No one should be at a disadvantage against cancer. The PSA Screening for HIM Act will help remove a major obstacle that can prevent those at high risk for the disease from getting the screening tests they need to find prostate cancer at the earliest, most treatable stage.”

    She added, “We urge the House and the Senate to pass this legislation to help reduce prostate cancer disparities and save more lives.”

    REFERENCES

    1. Kratzer TB, Mazzitelli N, Star J, et al. Prostate cancer statistics, 2025. CA: A Cancer Journal for Clinicians. 2025. doi:10.3322/caac.70028

    2. New ACS Prostate Cancer Statistics Report: Late-stage incidence rates continue to increase rapidly as mortality declines slow. News release. American Cancer Society. September 2, 2025. Accessed September 2, 2025. https://www.prnewswire.com/news-releases/new-acs-prostate-cancer-statistics-report-late-stage-incidence-rates-continue-to-increase-rapidly-as-mortality-declines-slow-302543895.html

    Continue Reading