The Role of Probiotics in Healing Burns and Skin Wounds; An Integrative Approach in the Context of Regenerative Medicine
Abstract
1. Introduction
2. Methods
2.1. Search Strategy and Data Sources
2.2. Eligibility Criteria and Assessment of Evidence
3. Epidemiological Aspects
4. The Pathophysiology of Burns and Tissue Healing
4.1. Local Burn Response
4.2. Systemic Response
4.3. Imbalances of Immunity in Burns
4.4. The Healing Process of Burn Wounds
4.5. The Role of Microbiome and the Impact on Regeneration
5. Classification of Burns and Conventional Therapeutic Approach
5.1. Classification According to the Depth of the Lesion [33]
- Grade I (superficial) burns: Affect exclusively the epidermal layer. Clinically, it is manifested by erythema, pain, and increased sensitivity to touch, without the formation of blisters. Healing occurs spontaneously in 3–6 days, without scarring, by rapid regeneration of basal keratinocytes. They are typical for excessive exposure to UV radiation (e.g., sunburn).
- Grade II burns (partially thick): Grade II superficial: Involve the epidermis and superficial portion of the papillary dermis. It is characterized by vesicles, intense erythema, and severe exudate pain. Healing occurs in 10–14 days, with complete restoration of the dermo–epidermal structure and minimal risk of scarring. Degree II deep: Affect up to the deep reticular dermis. The lesion areas are pale or marbled, the pain is reduced (due to damage to the nerve endings), and spontaneous healing is unlikely. In most cases, excision and covering with a cutaneous graft is required.
- Grade III burns (total thickness): Involve the complete destruction of the epidermis, dermis, and cutaneous appendages (pile follicles, sebaceous glands, sweat glands). The skin appears dry, white, brown, or charred and is insensitive to touch. Healing without surgery is impossible. Autologous cutaneous grafting is the therapeutic standard.
- Grade IV burns: Exceed the dermo–epidermal plane and affect the subcutaneous structures: fascia, muscle, tendons, bone. They are common in deep or explosive electrical injuries. The functional prognosis is severe, frequently requiring amputations or complex reconstructions.
5.2. Estimation of the Burn Extension
5.3. Topographic and Functional Classification
- Face—risk of deformity, microstomia, and ocular injury;
- Hands—high potential for joint contractures and loss of function;
- Perineum and genitals—increased infection risk, particularly challenging area to manage;
- Neck/throat region—prone to contracture formation that may impair airway and respiratory function;
- Large joints—burns involving biomechanical joints such as elbows, knees, or shoulders can lead to long-term functional impairment;
- Deep or extensive burns in these regions often necessitate early surgical excision and timely rehabilitation to preserve function and reduce aesthetic damage.
5.4. Conventional Therapeutic Approach
5.4.1. Minor Burns (Grade I and Superficial Grade II; TBSA Less than 10%) [36]
- Perform local antiseptic cleansing (e.g., chlorhexidine, povidone–iodine);
- Apply dressings: hydrocolloid, hydrogels, or silver-impregnated variants;
- Pain control: paracetamol or Non-Steroidal Anti-Inflammatory Drugs (NSAIDs);
- Avoid routine systemic antibiotics unless clinically indicated;
- Reassess clinically within 48–72 h to confirm healing trajectory.
5.4.2. Moderate to Severe Burns (Deep Grade II and Grade III; TBSA 15–20% in Adults) [37]
- Fluid resuscitation using the Parkland formula:
- Administer 50% of calculated volume in the first 8 h, remainder over the next 16 h;
- Only partial- and full-thickness burns count toward TBSA;
- Lactated Ringer’s solution is preferred unless contraindicated.
5.5. Limits of the Conventional Therapeutic Approach
6. Post-Burning Infections and Microbiotic Imbalances
6.1. Pathophysiological Mechanisms and Risk Factors of Post-Burn Infections
6.2. Post-Burn Sepsis and Multiorgan Dysfunction
6.3. Intestinal and Cutaneous Microbiota in the Context of Burns
- A significant reduction in beneficial commensal species (Bifidobacterium, Lactobacillus, Faecalibacterium prausnitzii);
- An increase in opportunistic pathogenic bacteria (Enterobacteriaceae, Clostridioides difficile, Pseudomonas aeruginosa);
- An increase in intestinal permeability accompanied by impaired intercellular cohesion, characterized by the downregulation of tight-junction proteins such as zonulin and occludin;
- An increase in the formation of the disease cutaneous dysbiosis with alteration of antimicrobial lipid production and inhibition of keratinocyte regeneration.
6.4. The Impact of Antibiotics on the Microbiome
6.5. Pathogens Involved in Nosocomial Infections
6.6. The Need for Complementary Therapeutic Strategies
7. The Role of Microbiome and Probiotics in Post-Burn Regeneration
7.1. The Cutaneous Microbiome and Epithelial Regeneration
7.2. Intestinal Microbiota and Remote Interactions
7.3. Experimental and Clinical Evidence on the Effects of Probiotics in Burns
7.3.1. Preclinical Evidence: Experimental Animal Models
7.3.2. Clinical Evidence
7.4. Probiotics Administration
7.4.1. Pharmaceutical Forms and Routes of Administration
7.4.2. Probiotics in Burn Care: Minimum Effective Doses
7.4.3. Co-Administration of Probiotics and Antibiotics
7.5. Current Challenges and Future Prospects
- The lack of validated clinical guidelines explicitly recommending certain strains;
- Compositional variability of commercial probiotic products without strict control standards;
- The need for large-sample randomized multicenter studies to validate clinical efficiency and long-term safety.
8. Synthesis of the Mechanisms of Action of Probiotics in Post-Burn Regeneration
- Immunomodulation;
- Tissue regeneration;
- Antimicrobial effect;
- Microbiotic balance and epithelial protection.
8.1. Modulation of Local and Systemic Immune Response
8.2. Tissue Regeneration and Angiogenesis
8.3. Direct and Indirect Antimicrobial Effect
8.4. Restoration of the Microbiotic Balance (Eubiosis)
9. Current Clinical Considerations and Future Therapeutic Perspectives
9.1. Indications and Profiles of Patients’ Candidates for Probiotic Therapy
- Patients with extensive burns (more than 15–20% TBSA) at increased risk of systemic infections and intestinal dysbiosis;
- Mechanically ventilated patients, enteral nutrients, with prolonged use of broad-spectrum antibiotics;
- Patients with leaky gut syndrome and markers of bacterial translocation;
- Patients with recurrent wound infections, colonization with Clostridium difficile;
- Patients in the subacute recovery phase, aimed at accelerating healing and reducing chronic inflammation.
9.2. Current Limitations and Challenges of Clinical Implementation
- Next-generation probiotics include fewer known strains with strong immunomodulatory effects, such as Faecalibacterium prausnitzii and Akkermansia muciniphila, with special applicability in chronic inflammation and epithelial regeneration [52].
- Postbiotics—bioactive substances produced by probiotics (SCFAs, peptides, polysaccharides, enzymes) administered directly without live microorganisms—offer increased stability and a higher safety profile [65].
- Synergistic bacterial-consortium therapy uses combinations of strains chosen for complementary action on immune response and tissue regeneration [66].
- Microbiome-personalized therapy tailors probiotics to the individual patient’s microbiotic profile determined by high-resolution metagenomics.
- Smart probiotic dressings—wound coverings impregnated with beneficial bacteria—create a microclimate favorable to regeneration while inhibiting pathogenic colonization [66].
9.3. Future Research Needs and Recommendations
- Large, multicenter randomized trials. Double-blind, placebo-controlled comparisons of well-defined probiotic (or synbiotic/postbiotic) formulations, powered for burn-specific primary outcomes (wound infection, graft take, time to closure/epithelialization) and key secondary outcomes (antibiotic days, length of stay, bacteremia/ventilator-associated pneumonia, organ-support-free days, patient-reported symptoms). Include pre-specified strata by burn severity (e.g., %TBSA, inhalation injury) and by route (topical vs. enteral).
- Integrated multi-omics. Longitudinal metagenomic, proteomic, and transcriptomic profiling of wound and gut samples to map effects on pathogen load, biofilm signatures, host inflammatory pathways, and tissue-repair programs using standardized sampling windows (baseline, day 3–5, pre-graft, post-graft, discharge).
- Pharmacokinetic/pharmacodynamic and formulation studies. Head-to-head evaluations of strains, doses and timing (early initiation ≤ 48 h vs. delayed), and routes (topical dressings vs. enteral) with measurement of viability at administration, persistence/clearance, local concentrations in the wound, and host-response markers; optimize delivery systems (e.g., hydrogels/smart dressings) for stability and controlled release.
- Safety surveillance. Burn-specific risk management with independent monitoring committees: systematic blood/wound cultures for potential translocation, adjudicated adverse events (including central line-associated events), whole-genome screening of candidate strains for resistance and virulence genes, and multicenter registries to detect rare harms.
- Economic evaluation. Prospective cost-effectiveness and budget-impact analyses capturing length of hospitalization, operating-room episodes, antibiotic consumption, readmissions, and quality-of-life metrics; include scenario and equity analyses relevant to low- and middle-income burn centers.
- Standardization and reporting. Adoption of a core outcome set for burn–microbiome trials, preregistered protocols, CONSORT-compliant reporting, clear strain identity (deposition numbers), explicit dose notation and viability both at end of shelf life and at administration, and transparent documentation of co-interventions (debridement, dressings, antibiotics).
- Regulatory translation. Early engagement with regulators on classification (live biotherapeutic vs. combination product), manufacturing quality, labeling and post-marketing pharmacovigilance to enable guideline inclusion once efficacy and safety are established.
10. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
Abbreviations
WHO | World Health Organization |
SIRS | systemic inflammatory response syndrome |
MRSA | Methicillin-resistant Staphylococcus aureus |
TBSA | total body surface area |
NSAIDs | Non-Steroidal Anti-Inflammatory Drugs |
MODS | multiple organ dysfunction syndrome |
SCFAs | short-chain fatty acids |
CFU | colony forming units |
PPI | anti-secretory drugs |
MDR | Multi-Drug Resistance |
IL-6 | Interleukina-6 |
IL-1 | Interleukina-1 |
IL-10 | Interleukina 10 |
IL-18 | Interleukina 18 |
PCR | Polymerase Chain Reaction |
IgA | Immunoglobulin A |
TGF | Transforming Growth Factor |
COL1A1 | Collagen TypeI Alpha 1Chain |
MMP1 | Matrix Metalloproteinase 1 |
MMP-9 | Matrix Metalloproteinase 9 |
ECM | Extracellular Matrix |
ICU | Intensive Care Unit |
Th1 | T helper 1 |
Th 17 | T helper 17 |
VEGF | Vascular Endothelial Growth Factor |
MAPK | Mitogen-Activated Protein Kinase |
PI3K/Akt | Phosphoinositide 3-Kinase / Protein Kinase B |
NF-κB | Nuclear Factor kappa-light-chain-enhancer of activated B cells |
LPS | Lipopolysaccharide |
References
- Church, D.; Elsayed, S.; Reid, O.; Winston, B.; Lindsay, R. Burn Wound Infections. Clin. Microbiol. Rev. 2006, 19, 403–434. [Google Scholar] [CrossRef]
- Salem, I.; Ramser, G.; Isham, C.; Ghannoum, M.H. The Gut Microbiome as a Major Regulator of the Gut-Skin Axis. Front. Microbiol. 2018, 9, 1459. [Google Scholar] [CrossRef]
- Khanh, V.K.S.; Syukur, S.; Purwati, E. Characterization of Lactic Acid Bacteria Isolated from Gambir with Potential Probiotic Properties. World Vet. J. 2025, 15, 1–8. [Google Scholar] [CrossRef]
- Fijan, S. Microorganisms with Claimed Probiotic Properties: An Overview of Recent Literature. Int. J. Environ. Res. Public Health 2014, 11, 4745–4767. [Google Scholar] [CrossRef] [PubMed]
- Gao, T.; Wang, X.; Li, Y.; Ren, F. The Role of Probiotics in Skin Health and Related Gut–Skin Axis: A Review. Nutrients 2023, 15, 3123. [Google Scholar] [CrossRef]
- Plaza-Díaz, J.; Ruiz-Ojeda, F.J.; Gil-Campos, M.; Gil, A. Mechanisms of Action of Probiotics. Adv. Nutr. 2019, 10, S49–S66. [Google Scholar] [CrossRef]
- Peral, M.C.; Rachid, M.M.; Gobbato, N.M.; Huaman Martinez, M.A.; Valdez, J.C. Interleukin-8 production by polymorphonuclear leukocytes from patients with chronic infected leg ulcers treated with Lactobacillus plantarum. Clin. Microbiol. Infect. 2010, 16, 281–286. [Google Scholar] [CrossRef]
- Herek, Ö.; Gökalan Kara, İ.; Kaleli, İ. Effects of Antibiotics and Saccharomyces boulardii on Bacterial Translocation in Burn Injury. Surg. Today 2004, 34, 256–260. [Google Scholar] [CrossRef]
- Partlow, J.; Blikslager, A.; Matthews, C.; Law, M.; Daniels, J.; Baker, R.; Labens, R. Effect of topically applied Saccharomyces boulardii on the healing of acute porcine wounds: A preliminary study. BMC Res. Notes 2016, 9, 210. [Google Scholar] [CrossRef]
- Swanson, K.S.; Gibson, G.R.; Hutkins, R.; Reimer, R.A.; Reid, G.; Verbeke, K.; Scott, K.P.; Holscher, H.D.; Azad, M.B.; Delzenne, N.M.; et al. The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of synbiotics. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 687–701. [Google Scholar] [CrossRef]
- Taverniti, V.; Guglielmetti, S. The immunomodulatory properties of probiotic microorganisms beyond their viability (ghost probiotics: Proposal of paraprobiotic concept). Genes Nutr. 2011, 6, 261–274. [Google Scholar] [CrossRef] [PubMed]
- Hill, D.A.; Artis, D. Intestinal bacteria and the regulation of immune cell homeostasis. Annu. Rev. Immunol. 2010, 28, 623–667. [Google Scholar] [CrossRef]
- Sanders, M.E.; Merenstein, D.J.; Reid, G.; Gibson, G.R.; Rastall, R.A. Probiotics and prebiotics in intestinal health and disease: From biology to the clinic. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 605–616. [Google Scholar] [CrossRef] [PubMed]
- Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef]
- Suez, J.; Zmora, N.; Zilberman-Schapira, G.; Mor, U.; Dori-Bachash, M.; Bashiardes, S.; Zur, M.; Regev-Lehavi, D.; Ben-Zeev Brik, R.; Federici, S.; et al. Post-Antibiotic Gut Mucosal Microbiome Reconstitution Is Impaired by Probiotics and Improved by Autologous FMT. Cell 2018, 174, 1406–1423.e16. [Google Scholar] [CrossRef]
- World Health Organization. Burns. 2023. Available online: https://www.who.int/news-room/fact-sheets/detail/burns (accessed on 25 July 2025).
- Peck, M.D. Epidemiology of Burns throughout the World. Part I: Distribution and Risk Factors. Burns 2011, 37, 1087–1100. [Google Scholar] [CrossRef]
- Mayes, T.; Gottschlich, M.M.; James, L.P.; Allgeier, C.; Weitz, J.; Kagan, R.J. Clinical Safety and Efficacy of Probiotic Administration Following Burn Injury. J. Burn Care Res. 2015, 36, 92–99. [Google Scholar] [CrossRef]
- EuroSafe. Injuries in the European Union 2009–2018. 2021. Available online: https://www.eurosafe.eu.com/uploads/inline-files/Report%20Injuries%20in%20EU%202009-2018%20FINAL%20SEPT%2021_pdf (accessed on 25 July 2025).
- Zhang, P.; Zou, B.; Liou, Y.; Huang, C. The pathogenesis and diagnosis of sepsis post burn injury. Burn. Trauma 2021, 9, tkaa047. [Google Scholar] [CrossRef]
- National DRG Center/School of Public Health, Romania. Epidemiology of hospitalized burns in Romania: A 10-year study on 92333 patients. Burns 2020. Data sourced from Romanian national DRG database, 2006–2015.. [Google Scholar]
- StatPearls/Thermal Burns Overview. Thermal Burns: Overview, Pathophysiology, Quantifying Burn Sevrity. 2025. Available online: https://www.ncbi.nlm.nih.gov/books/NBK430773/ (accessed on 30 May 2025).
- Hop, M.J.; Polinder, S.; van der Vlies, C.H.; Middelkoop, E.; van Baar, M.E. Costs of burn care: A systematic review. Wound Repair Regen. 2014, 22, 436–450. [Google Scholar] [CrossRef]
- Jeschke, M.G.; Chinkes, D.L.; Finnerty, C.C.; Kulp, G.; Suman, O.E.; Norbury, W.B.; Branski, L.K.; Gauglitz, G.G.; Mlcak, R.P.; Herndon, D.N. Pathophysiologic response to severe burn injury. Ann. Surg. 2008, 248, 387–400. [Google Scholar] [CrossRef]
- Vinaik, R.; Salsali, H.; Jeschke, M.G.; Patsouris, D. Advances in Understanding Systemic Inflammation and Immunity in Burn Injuries. Int. J. Mol. Sci. 2023, 24, 3749. [Google Scholar] [CrossRef]
- Wang, Z.; Xiao, G.; Yao, Y.; Guo, S.; Lu, K.; Sheng, Z. The role of bifidobacteria in gut barrier function after thermal injury in rats. J. Trauma 2006, 61, 650–657. [Google Scholar] [CrossRef] [PubMed]
- Huang, Z.; Huang, Y.; Chen, J.; Tang, Z.; Chen, Y.; Liu, H.; Huang, M.; Qing, L.; Li, L.; Wang, Q.; et al. The role and therapeutic potential of gut microbiome in severe burn. Front. Cell. Infect. Microbiol. 2022, 12, 1–16. [Google Scholar] [CrossRef] [PubMed]
- Ward, J.A.; Phillips, G.; Martin, N.A.J. Accurately identifying blast injuries in civilian burn practice. Burns 2018, 44, 1610–1611. [Google Scholar] [CrossRef]
- Magnotti, L.J.; Deitch, E.A. Burns, bacterial translocation, gut barrier function, and failure. J. Burn Care Rehabil. 2005, 26, 383–391. [Google Scholar] [CrossRef]
- Wang, X.; Yang, J.; Tian, F.; Zhang, L.; Lei, Q.; Jiang, T.; Zhou, J.; Yuan, S.; Wang, J.; Feng, Z.; et al. Gut microbiota trajectory in patients with severe burn: A time series study. J. Crit. Care 2017, 42, 310–316. [Google Scholar] [CrossRef]
- Jeschke, M.G.; van Baar, M.E.; Choudhry, M.A.; Chung, K.K.; Gibran, N.S.; Logsetty, S. Burn injury. Nat. Rev. Dis. Primers 2020, 6, 11. [Google Scholar] [CrossRef]
- Özdemir, B.; Akıncı, E.; But, A.; Kazancioğlu, S.; Yastı, A.; Yüksek, Y.N.; Sözen, İ.; Bodur, H. Bloodstream Infections in Severe Burn Patients: Epidemiology, Microbiology, Laboratory Features, and Risk Factors Associated with Mortality. Mediterr. J. Infect. Microbes Antimicrob. 2022, 11, 47. [Google Scholar] [CrossRef]
- Classification of Burns. Available online: https://www.nationwidechildrens.org/conditions/health-library/classification-of-burns (accessed on 20 June 2025).
- García-Ballesteros, D.I.; Rivera-Martínez, D.D.C.; García-Pérez, M.M.; Valdés-Flores, E.; Castro-Govea, Y.; Chacón-Moreno, H.J. Evaluation and Optimization of the Wallace Rule of Nines for the Estimation of Total Body Surface Area in Obese and Nonobese Populations. J. Emerg. Med. 2023, 65, e320–e327. [Google Scholar] [CrossRef]
- Giretzlehner, M.; Ganitzer, I.; Haller, H. Technical and Medical Aspects of Burn Size Assessment and Documentation. Medicina 2021, 57, 242. [Google Scholar] [CrossRef]
- Markiewicz-Gospodarek, A.; Kozioł, M.; Tobiasz, M.; Baj, J.; Radzikowska-Büchner, E.; Przekora, A. Burn Wound Healing: Clinical Complications, Medical Care, Treatment, and Dressing Types—The Current State of Knowledge for Clinical Practice. Int. J. Environ. Ment. Res. Public Health 2022, 19, 1338. [Google Scholar] [CrossRef] [PubMed]
- Al Jebawi, K.; Toami, J.; Ayash, A.; Salha, R.; Al Shoufy, M.; Nasrallah, J.; Danaf, N.; El Ghawi, L.; Msheik, L.; Hamdar, H. Probiotics: A Promising Ally in Burn Treatment. ASIDE Int. Med. 2025, 1, 10–17. [Google Scholar] [CrossRef]
- Martín, R.; Miquel, S.; Ulmer, J.; Kechaou, N.; Langella, P.; Bermúdez-Humarán, L.G. Role of commensal and probiotic bacteria in human health: A focus on inflammatory bowel disease. Microb. Cell Factories 2013, 12, 71. [Google Scholar] [CrossRef] [PubMed]
- Mohajeri, H.M.; Brummer, R.J.M.; Rastall, R.A.; Weersma, R.K.; Harmsen, H.J.M.; Faas, M.; Eggersdorfer, M. The role of the microbiome for human health: From basic science to clinical applications. Eur. J. Nutr. 2018, 57, 1–14. [Google Scholar] [CrossRef]
- Hemarajata, P.; Versalovic, J. Effects of probiotics on gut microbiota: Mechanisms of intestinal immunomodulation and neuromodulation. Ther. Adv. Gastroenterol. 2013, 6, 39–51. [Google Scholar] [CrossRef]
- Kober, M.; Bowe, W.P. The effect of probiotics on immune regulation, acne, and photoaging. Int. J. Women’s Dermatol. 2015, 1, 85–89. [Google Scholar] [CrossRef]
- Corrêa-Oliveira, R.; Fachi, J.L.; Vieira, A.; Sato, F.T.; Vinolo, M.A.R. Regulation of immune cell function by short-chain fatty acids. Clin. Transl. Immunol. 2016, 5, e73. [Google Scholar] [CrossRef]
- Vinolo, M.A.R.; Rodrigues, H.G.; Nachbar, R.T.; Curi, R. Regulation of Inflammation by Short Chain Fatty Acids. Nutrients 2011, 3, 858–876. [Google Scholar] [CrossRef]
- Earley, Z.M.; Akhtar, S.; Green, S.J.; Naqib, A.; Khan, O.; Cannon, A.R.; Hammer, A.M.; Morris, N.L.; Li, X.; Eberhardt, J.M.; et al. Burn Injury Alters the Intestinal Microbiome and Increases Gut Permeability and Bacterial Translocation. PLoS ONE 2015, 10, e0129996. [Google Scholar] [CrossRef]
- Buffie, C.G.; Pamer, E.G. Microbiota-mediated colonization resistance against intestinal pathogens. Nat. Rev. Immunol. 2013, 13, 790–801. [Google Scholar] [CrossRef]
- Lee, Y.M.; Jo, K.; Kim, S.Y.; Seo, C.-S.; Son, E.; Kim, A.; Kim, D.-S. Yeokwisan: Standardised Herbal Formula Enhancing Gastric Mucosal Protection Against Gastric Ulcers in Mice, a Preclinical Study. Pharmaceuticals 2025, 18, 44. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Maitz, J.; Merlino, J.; Rizzo, S.; McKew, G.; Maitz, P. Burn wound infections microbiome and novel approaches using therapeutic microorganisms in burn wound infection control. Adv. Drug Deliv. Rev. 2023, 196, 114769. [Google Scholar] [CrossRef] [PubMed]
- Mills, J.P.; Rao, K.; Young, V.B. Probiotics for prevention of Clostridium difficile infection. Curr. Opin. Gastroenterol. 2018, 34, 3–10. [Google Scholar] [CrossRef] [PubMed]
- Meenakshi, S.; Santhanakumar, R. The role of probiotics as wound healers: An overall view. J. Wound Care 2023, 32, 318–328. [Google Scholar] [CrossRef]
- Tang, Q.; Xue, N.; Ding, X.; Tsai, K.H.Y.; Hew, J.J.; Jiang, R.; Huang, R.; Cheng, X.; Ding, X.; Cheng, Y.Y.; et al. Role of wound microbiome, strategies of microbiota delivery system and clinical management. Adv. Drug Deliv. Rev. 2023, 192, 114671. [Google Scholar] [CrossRef]
- Ríos-Covián, D.; Ruas-Madiedo, P.; Margolles, A.; Gueimonde, M.; de los Reyes-Gavilán, C.G.; Salazar, N. Intestinal Short Chain Fatty Acids and Their Link with Diet and Human Health. Front. Microbiol. 2016, 7, 185. [Google Scholar] [CrossRef]
- Machado, P.; Ribeiro, F.N.; Giublin, F.C.W.; Mieres, N.G.; Tonin, F.S.; Pontarolo, R.; Sari, M.H.M.; Lazo, R.E.L.; Ferreira, L.M. Next-Generation Wound Care: A Scoping Review on Probiotic, Prebiotic, Synbiotic, and Postbiotic Cutaneous Formulations. Pharmaceuticals 2025, 18, 704. [Google Scholar] [CrossRef]
- Valdéz, J.C.; Peral, M.C.; Rachid, M.; Santana, M.; Perdigón, G. Interference of Lactobacillus plantarum with Pseudomonas aeruginosa in vitro and in infected burns: The potential use of probiotics in wound treatment. Clin. Microbiol. Infect. 2005, 11, 472–479. [Google Scholar] [CrossRef]
- Shimizu, K.; Yamada, T.; Ogura, H.; Mohri, T.; Kiguchi, T.; Fujimi, S.; Asahara, T.; Yamada, T.; Ojima, M.; Ikeda, M.; et al. Synbiotics modulate gut microbiota and reduce enteritis and ventilator-associated pneumonia in patients with sepsis: A randomized controlled trial. Crit. Care 2018, 22, 239. [Google Scholar] [CrossRef]
- Manzanares, W.; Lemieux, M.; Langlois, P.L.; Wischmeyer, P.E. Probiotic and symbiotic therapy in critical illness: A systematic review and meta-analysis. Crit. Care 2016, 20, 262. [Google Scholar] [CrossRef]
- Baral, K.C.; Bajracharya, R.; Lee, S.H.; Han, H.K. Advancements in the Pharmaceutical Applications of Probiotics: Dosage Forms and Formulation Technology. Int. J. Nanomed. 2021, 16, 7535–7556. [Google Scholar] [CrossRef]
- Yin, Z.; Qiu, Y.; Han, Y.; Li, K. Topical probiotics in wound care: A review of effects, mechanisms, and applications. Interdiscip. Nurs. Res. 2024, 3, 63–71. [Google Scholar] [CrossRef]
- Tandon, D.; Haque, M.M.; Gote, M.; Jain, M.; Bhaduri, A.; Dubey, A.K.; Mande, S.S. A prospective randomized, double-blind, placebo-controlled, dose-response relationship study to investigate efficacy of fructo-oligosaccharides (FOS) on human gut microflora. Sci. Rep. 2019, 9, 5473. [Google Scholar] [CrossRef]
- International Scientific Association for Probiotics and Prebiotics (ISAPP). Probiotic Use Along-side Antibiotics: A Guide to Clinical FAQs. In ISAPP Clinical Briefs; International Scientific Association for Probiotics and Prebiotics (ISAPP): 2024. Available online: https://isappscience.org/wp-content/uploads/2024/12/ISAPP-Clinician-resource-probiotic-use-alongside-antibiotics.pdf (accessed on 25 July 2025).
- Osuka, A.; Shigeno, A.; Matsuura, H.; Onishi, S.; Yoneda, K. Systemic immune response of burns from the acute to chronic phase. Acute Med. Surg. 2024, 11, e976. [Google Scholar] [CrossRef] [PubMed]
- Ishii, S.; Kanno, E.; Tanno, H.; Kurosaka, S.; Shoji, M.; Imai, T.; Yamaguchi, K.; Kotsugai, K.; Niiyama, M.; Kurachi, H.; et al. Cutaneous wound healing promoted by topical heat-killed Lactobacillus plantarum KB131 and possible contribution of CARD9-mediated signaling. Sci. Rep. 2023, 13, 15917. [Google Scholar] [CrossRef] [PubMed]
- Han, N.; Jia, L.; Su, Y.; Du, J.; Guo, L.; Luo, Z.; Liu, Y. Lactobacillus reuteri extracts promoted wound healing. Stem Cell Res. Ther. 2019, 10, 243. [Google Scholar] [CrossRef] [PubMed]
- Fernández-Guarino, M.; Hernández-Bule, M.L.; Bacci, S. Cellular and Molecular Processes in Wound Healing. Biomedicines 2023, 11, 2526. [Google Scholar] [CrossRef]
- Dicks, L.M.T.; Botes, M. Probiotic lactic acid bacteria in the gastro-intestinal tract: Health benefits, safety and mode of action. Benef. Microbes 2010, 1, 11–29. [Google Scholar] [CrossRef]
- Nagpal, R.; Kumar, A.; Kumar, M.; Behare, P.V.; Jain, S.; Yadav, H. Probiotics, their health benefits and applications for developing healthier foods: A review. FEMS Microbiol. Lett. 2012, 334, 1–15. [Google Scholar] [CrossRef]
- El-Mesery, M.; Saleh, S. Impact of topical application of Lactobacillus on burn wound healing: A pilot study. Burn. Trauma 2022, 10, tkac011. [Google Scholar] [CrossRef]
- Peral, M.C.; Huaman Martinez, M.A.; Valdez, J.C. Bacteriotherapy with Lactobacillus plantarum in burns. Int. Wound J. 2009, 6, 73–81. [Google Scholar] [CrossRef]
- El-Ghazely, M.H.; Mahmoud, W.H.; Atia, M.A.; Eldip, E.M. Effect of probiotic administration in the therapy of pediatric thermal burn. Ann. Burns Fire Disasters 2016, 29, 268–272. [Google Scholar]
Product/Strain | Typical Daily Dose | Range | Prebiotic | Key Notes |
---|---|---|---|---|
Minimum effective (general) | ≥ 1 × 109 CFU/day | 109–1011 CFU/day | — | severity; in burn care start within 24–48 h. |
Lactobacillus rhamnosus GG (LGG) | 1–2 × 1010 CFU/day | 109–2 × 1010 CFU/day | Optional (inulin) | Well studied; prefer gastro-resistant oral forms. |
Saccharomyces boulardii | 250–500 mg/day (≈ 5–10 × 109 CFU) | 5 × 109–1 × 1010 CFU/day | Not required | Avoid in severe immunosuppression or with central venous catheters. |
Bifidobacterium longum | 1 × 109 CFU/day | 109–1010 CFU/day | Often inulin | Synbiotic pairing may enhance engraftment. |
Synbiotic formulas | 109–1011 CFU/day | — | 2–4 g/day (inulin) | Match prebiotic to strain; verify |
Severe Burn Injury/Immune Dysregulation | Probiotic-Mediated Mechanisms | Restored Immune Balance |
---|---|---|
Hyper-inflammatory phase - IL-6 ↑ - Oxidative stress - Complement activation | - Reduction in pro-inflammatory cytokines (IL-6, TNF-α) - Induction of anti-inflammatory cytokines (IL-10) | - Attenuation of excessive systemic inflammation |
Immunoparalysis phase - IL-10 ↑ - MHC-II on monocytes ↓ - T-cell suppression | - Activation of tolerogenic dendritic cells - T-cell polarization toward Treg (↓Th1/Th17) - Regulation of co-stimulatory molecules - Restoration of monocyte/macrophage function | - Rebalanced innate and adaptive responses - Functional immune recovery |
Barrier dysfunction - Impaired mucosal immunity | - Increased secretory IgA production - Strengthening of mucosal barrier | - Improved barrier integrity - Enhanced resistance to infection |
Subgroup | Outcome | Probiotic (n/N, %) | Control (n/N, %) | ∆ pp (95% CI) | RR (95% CI) |
---|---|---|---|---|---|
Delayed II° (infected) | Composite * | 10/14 (71.4) | 11/15 (73.3) | −1.9 (−34.5–30.7) | 0.97 (0.62–1.53) |
Early III° (non-infected) | Granulation for grafting | 10/12 (83.3) | 11/13 (84.6) | −1.3 (−30.1–27.5) | 0.98 (0.70–1.39) |
Early III° (non-infected) | Graft take | 9/10 (90.0) | 10/11 (90.9) | −0.9 (−26.1–24.3) | 0.99 (0.75–1.31) |
Early III° (non-infected) | Complete healing | 9/12 (75.0) | 10/13 (76.9) | −1.9 (−35.5–31.6) | 0.97 (0.63–1.52) |
Delayed III° (infected) | Composite † | 10/12 (83.3) | 10/14 (71.4) | +11.9 (−19.8–43.6) | 1.17 (0.77–1.77) |
Delayed III° (infected) | Graft take | 9/10 (90.0) | 9/10 (90.0) | 0.0 (−26.3–26.3) | 1.00 (0.75–1.34) |
Delayed III° (infected) | Complete healing | 9/12 (75.0) | 9/14 (64.3) | +10.7 (−24.4–45.8) | 1.17 (0.70–1.94) |
Notes: ∆ pp = absolute difference in percentage points (probiotic minus control); RR = risk ratio. * Composite = bacterial load < 105/g + granulation + healing. † Composite = bacterial load < 105/g + granulation. Subgroup sizes ∼12–15 patients; Source: Peral et al. [67]. |
Key Point | Brief Mechanism/Effect | Ref. |
---|---|---|
Burn injury triggers systemic alterations with gut dysbiosis | Systemic changes that influence cutaneous | [43] |
Intestine hosts ∼70–80% of host immune activity | Microbiota modulates systemic inflammation, vascular barrier function, and mesenchymal stem cells | [43] |
Gut–skin axis | Microbial metabolites link gut signals to skin healing responses | [52] |
SCFAs (butyrate, propionate, acetate) | Keratinocyte migration/proliferation ↑; Angiogenesis↑ collagen sinthesis ↑ | [52] |
Probiotics elevating SCFA in burn models | Systemic inflammation ↓; bacterial translocation; re-epithelization↑ | [53] |
Burn Severity/Population | Intervention | Main Outcomes |
---|---|---|
Pediatric moderate–severe burns | Oral probiotic, 1010 CFU/dose, twice daily | Feasible integration into standard burn care; safe and well tolerated; potential reduction in infections and intestinal dysbiosis. 1 |
Pediatric burns (n = 80), single-center | Probiotics as adjuvant therapy | Indications of immunomodulatory effects; limited by small sample size and lack of multicenter validation. 1 |
Adults with second- and third-degree burns | Topical Lactobacillus plantarum vs. 1% silver sulfadiazine | Comparable or superior efficacy: lower bacterial load, improved granulation, complete healing in 75–77% of cases; no invasive infections; well tolerated. 2 |
Infected, delayed-healing burns | Topical L. plantarum | Reduced bacterial colonization; improved granulation (83% vs. 71% in controls); higher complete healing rate (75% vs. 64%). 2 |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Ambrose, L.; Dinu, C.A.; Gurau, G.; Maftei, N.-M.; Matei, M.N.; Hincu, M.-A.; Radu, M.; Mehedinti, M.-C. The Role of Probiotics in Healing Burns and Skin Wounds; An Integrative Approach in the Context of Regenerative Medicine. Life 2025, 15, 1434. https://doi.org/10.3390/life15091434
Ambrose L, Dinu CA, Gurau G, Maftei N-M, Matei MN, Hincu M-A, Radu M, Mehedinti M-C. The Role of Probiotics in Healing Burns and Skin Wounds; An Integrative Approach in the Context of Regenerative Medicine. Life. 2025; 15(9):1434. https://doi.org/10.3390/life15091434
Chicago/Turabian StyleAmbrose, Lenuta, Ciprian Adrian Dinu, Gabriela Gurau, Nicoleta-Maricica Maftei, Madalina Nicoleta Matei, Maria-Andrada Hincu, Marius Radu, and Mihaela-Cezarina Mehedinti. 2025. "The Role of Probiotics in Healing Burns and Skin Wounds; An Integrative Approach in the Context of Regenerative Medicine" Life 15, no. 9: 1434. https://doi.org/10.3390/life15091434
APA StyleAmbrose, L., Dinu, C. A., Gurau, G., Maftei, N.-M., Matei, M. N., Hincu, M.-A., Radu, M., & Mehedinti, M.-C. (2025). The Role of Probiotics in Healing Burns and Skin Wounds; An Integrative Approach in the Context of Regenerative Medicine. Life, 15(9), 1434. https://doi.org/10.3390/life15091434