Nothing Special   »   [go: up one dir, main page]

You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (970)

Search Parameters:
Keywords = chronic wound healing

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
12 pages, 1213 KiB  
Review
Platelet-Rich Plasma in Diabetic Foot Ulcer Healing: Contemplating the Facts
by Jacob Smith and Vikrant Rai
Int. J. Mol. Sci. 2024, 25(23), 12864; https://doi.org/10.3390/ijms252312864 (registering DOI) - 29 Nov 2024
Abstract
Diabetic foot ulcers (DFUs), debilitating complication of diabetes, often lead to amputation even in the presence of current advanced treatment for DFUs. Platelet-rich plasma (PRP) containing growth factors and other proteins has been suggested as a potent therapeutic in promoting DFU healing. PRP [...] Read more.
Diabetic foot ulcers (DFUs), debilitating complication of diabetes, often lead to amputation even in the presence of current advanced treatment for DFUs. Platelet-rich plasma (PRP) containing growth factors and other proteins has been suggested as a potent therapeutic in promoting DFU healing. PRP is safe and effective in improving the DFU healing rate, decreasing healing time, and making chronic wounds viable for treatment. Though PRP is safe and effective in promoting DFU healing, there are inconsistencies in clinical outcomes. These varying results may be due to various concentrations of PRP being used. Most studies report dosage and timing, but none have reported the concentration of various factors. This is important, as the concentration of factors in PRP can vary significantly with each preparation and may directly impact the healing outcome. This critical review discusses the limiting factors and issues related to PRP therapy and future directives. A systematic search of PubMed and Google Scholar was performed with keywords including diabetic foot ulcer, ulcer healing, platelet-rich plasma, DFU treatment, and PRP limitations and efficacy, alone or in combination, to search the related articles. The articles describing DFU and the use of PRP in DFU healing were included. The existing literature suggests that PRP is effective and safe for promoting DFU healing, but larger clinical trials are needed to improve clinical outcomes. There is a need to consider multiple factors including the role of epigenetics, lifestyle modification, and the percentage composition of each constituent in PRP. Full article
(This article belongs to the Section Bioactives and Nutraceuticals)
Show Figures

Figure 1

Figure 1
<p>Preparation of platelet-rich plasma. The first centrifugation separates the whole blood into three layers: 1. red blood cell layer (RBC), 2. buffy coat (BC) containing white blood cells and most of the platelets, and 3. platelet-poor plasma (PPP) layer. The second centrifugation of BC and PPP separates all platelets in platelet-rich plasma (PRP) layer and PPP.</p>
Full article ">Figure 2
<p>Molecular and cellular role supplemented by platelet-rich plasma (PRP) when administered to wound. The growth factors stored in platelets are secreted and stimulate the proliferation and migration of fibroblasts, keratinocytes, vascular smooth muscle cells (VSMCs), and endothelial cells toward the wound area. These cells and their interaction in-between and with immune cells help in promoting wound healing by suppressing inflammation and promoting tissue regeneration. PDGF—platelet-derived growth factor, EGF—epidermal growth factor, FGF—fibroblast growth factor, VEGF—vascular endothelial growth factor, IGF—insulin-like growth factor, TGF-β—transforming growth factor-beta, CD34—cluster of differentiation 34, HSP47—heat shock protein 47, SFA—fibroblast-specific surface antigen, SMA—smooth muscle actin, ECM—extracellular matrix, and IL-interleukin.</p>
Full article ">
22 pages, 1203 KiB  
Review
Aberrances of the Wound Healing Process: A Review
by Montserrat Fernandez-Guarino, Jorge Naharro-Rodriguez and Stefano Bacci
Cosmetics 2024, 11(6), 209; https://doi.org/10.3390/cosmetics11060209 - 28 Nov 2024
Viewed by 225
Abstract
Wound healing is a complex biological process that can lead to chronic wounds, keloids, and hypertrophic scars when disrupted. Chronic wounds result from a prolonged inflammatory phase and impaired re-epithelialization. Keloids are characterized by excessive collagen deposition beyond the original wound boundaries, driven [...] Read more.
Wound healing is a complex biological process that can lead to chronic wounds, keloids, and hypertrophic scars when disrupted. Chronic wounds result from a prolonged inflammatory phase and impaired re-epithelialization. Keloids are characterized by excessive collagen deposition beyond the original wound boundaries, driven by persistent inflammation and fibroblast hyperactivity. Hypertrophic scars, on the other hand, are confined to the wound edges and are caused by an imbalance in collagen synthesis and degradation, typically resolving over time. The therapeutic approach to wound healing impairment involves a range of strategies, including non-invasive (which focus on supporting the natural healing process), minimally invasive, and aggressive interventions (such as surgical approach, often reserved for severe or refractory cases). Emerging therapies, including stem cell treatments and botulinum toxin injections, offer new hope for improving outcomes in patients with wound healing impairments. This review highlights the distinct mechanisms underlying chronic wounds, keloids, and hypertrophic scars and discusses their respective therapeutic approaches, focusing on both established and emerging therapies. Understanding these mechanisms is crucial for optimizing treatment strategies and improving patient outcomes. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Typical image of a venous ulcer in the pretibial area, secondary to chronic venous insufficiency and cutaneous sclerosis, and (<b>B</b>) typical clinical aspect of diabetic foot ulcers, provided by Dr. Almudena Cecilia-Matilla, Diabetic Foot Unit, Angiology and Vascular Surgery Department. Ramón y Cajal University Hospital.</p>
Full article ">Figure 2
<p>Image of a keloid in the ear, which appears typically after getting an ear piercing.</p>
Full article ">Figure 3
<p>Flowchart depicting available therapies for keloid management and their progression from more conservative to more aggressive.</p>
Full article ">
26 pages, 8645 KiB  
Review
The Immune-Centric Revolution Translated into Clinical Application: Peripheral Blood Mononuclear Cell (PBMNC) Therapy in Diabetic Patients with No-Option Critical Limb-Threatening Ischemia (NO-CLTI)—Rationale and Meta-Analysis of Observational Studies
by Laura Rehak, Laura Giurato, Matteo Monami, Marco Meloni, Alessia Scatena, Andrea Panunzi, Giada Maria Manti, Carlo Maria Ferdinando Caravaggi and Luigi Uccioli
J. Clin. Med. 2024, 13(23), 7230; https://doi.org/10.3390/jcm13237230 - 28 Nov 2024
Viewed by 324
Abstract
Chronic limb-threatening ischemia (CLTI), the most advanced form of peripheral arterial disease (PAD), is the comorbidity primarily responsible for major lower-limb amputations, particularly for diabetic patients. Autologous cell therapy has been the focus of efforts over the past 20 years to create non-interventional [...] Read more.
Chronic limb-threatening ischemia (CLTI), the most advanced form of peripheral arterial disease (PAD), is the comorbidity primarily responsible for major lower-limb amputations, particularly for diabetic patients. Autologous cell therapy has been the focus of efforts over the past 20 years to create non-interventional therapeutic options for no-option CLTI to improve limb perfusion and wound healing. Among the different available techniques, peripheral blood mononuclear cells (PBMNC) appear to be the most promising autologous cell therapy due to physio-pathological considerations and clinical evidence, which will be discussed in this review. A meta-analysis of six clinical studies, including 256 diabetic patients treated with naive, fresh PBMNC produced via a selective filtration point-of-care device, was conducted. PBMNC was associated with a mean yearly amputation rate of 15.7%, a mean healing rate of 62%, and a time to healing of 208.6 ± 136.5 days. Moreover, an increase in TcPO2 and a reduction in pain were observed. All-cause mortality, with a mean rate of 22.2% and a yearly mortality rate of 18.8%, was reported. No serious adverse events were reported. Finally, some practical and financial considerations are provided, which point to the therapy’s recommendation as the first line of treatment for this particular and crucial patient group. Full article
(This article belongs to the Section Endocrinology & Metabolism)
Show Figures

Figure 1

Figure 1
<p>Angiography pre- and post-PBMNC implants. (<b>A</b>) Pre- (Time 0) and post-14 weeks (Time 1) after a PBMNC implant in diabetic patients with no-option CLTI: the sprouting of new vasa interspersed through all of the foot (unpublished data from the University of Tor Vergata, Rome). (<b>B</b>) (i) Patient after unsuccessful PTA (TcPO2 &lt; 30 mmHg) therapy showing the typical “desert” foot condition; (ii) patient after 2 months of PB-MNC therapy showing collateral vascular remodeling (Panunzi et al. [<a href="#B20-jcm-13-07230" class="html-bibr">20</a>]).</p>
Full article ">Figure 2
<p>Morphological aspects and an immunohistochemical analysis from biopsies taken pre- (T0) and 30 days post-PBMNC implant (T1) in a diabetic foot wound area (M1 (CD38+) and M2 (CD163+)). At the top (first row), hematoxylin and eosin (H&amp;E) staining shows an inflamed diabetic wound due to ulceration at Time 0 and an important area of regenerated tissue at Time 1. In the second row, at T0, CD38+ immunostaining highlights a large M1 macrophage inflammatory infiltration (10×), and at T1, the quantity and intensity of CD38+ staining for M1 were both reduced (10×). In the third row, at T0, rare M2 macrophages were positive for the CD163 marker (10×), whereas the amount of CD163+ M2 cells significantly increased at Time 1 (10×). Courtesy of Manuela Montanaro (Department Biomedicine and Prevention) and Alessandro Mauriello (Department Experimental Medicine-University of Tor Vergata Roma) (Unpublished data Policlinico Tor Vergata Roma). Immunohistochemistry and immunofluorescence methods are described in the Materials and Methods section.</p>
Full article ">Figure 3
<p>Macrophage polarization quantification expressed in the number of positive cell/area (CD38+ M1 and CD163+ M2) measured in immunohistochemical samples from a wound area before (T0) and after 30 days (T1) a PBMNC implant in three representative patients (<span class="html-italic">n</span> = 3). Courtesy of Manuela Montanaro (Department Biomedicine and Prevention) and Alessandro Mauriello (Department Experimental Medicine- University of Tor Vergata Roma); unpublished data, Policlinico Tor Vergata Roma. Immunohistochemistry and immunofluorescence methods are described in the Materials and Methods section.</p>
Full article ">Figure 4
<p>Clinical case of diabetic patient with no-option CLTI before and after three implants of PBMNCs. Clinical pictures: from the upper left at the time of the first PBMNC implant to the lower right six months later. Pictures were taken at the baseline (<b>A</b>), after the first, second, and third PBMNC implants (<b>B</b>–<b>D</b>), one month after the final PBMNC implant (<b>E</b>), and at a 6-month follow-up (<b>F</b>).</p>
Full article ">Figure 5
<p>Major amputation with PBMNCs in all included studies (<b>A</b>), yearly rate and in case-control studies (<b>B</b>), at the endpoint. Reported references are Scatena et al [<a href="#B19-jcm-13-07230" class="html-bibr">19</a>], Panunzi et al, [<a href="#B20-jcm-13-07230" class="html-bibr">20</a>], Ragghianti et al [<a href="#B21-jcm-13-07230" class="html-bibr">21</a>], Meloni et al [<a href="#B22-jcm-13-07230" class="html-bibr">22</a>], Persiani et al [<a href="#B24-jcm-13-07230" class="html-bibr">24</a>], De Angelis et al [<a href="#B26-jcm-13-07230" class="html-bibr">26</a>].</p>
Full article ">Figure 6
<p>Ulcer healing rate with PBMNCs in all included studies. Reported references are Scatena et al [<a href="#B19-jcm-13-07230" class="html-bibr">19</a>], Panunzi et al, [<a href="#B20-jcm-13-07230" class="html-bibr">20</a>], Ragghianti et al [<a href="#B21-jcm-13-07230" class="html-bibr">21</a>], Meloni et al [<a href="#B22-jcm-13-07230" class="html-bibr">22</a>].</p>
Full article ">Figure 7
<p>All-cause mortality (yearly rate) with PBMNCs in all included studies. Reported references are Scatena et al [<a href="#B19-jcm-13-07230" class="html-bibr">19</a>], Panunzi et al, [<a href="#B20-jcm-13-07230" class="html-bibr">20</a>], Ragghianti et al [<a href="#B21-jcm-13-07230" class="html-bibr">21</a>], Meloni et al [<a href="#B22-jcm-13-07230" class="html-bibr">22</a>], Persiani et al [<a href="#B24-jcm-13-07230" class="html-bibr">24</a>], De Angelis et al [<a href="#B26-jcm-13-07230" class="html-bibr">26</a>].</p>
Full article ">
20 pages, 952 KiB  
Review
Critical Analysis of Cytoplasmic Progression of Inflammatory Signaling Suggests Potential Pharmacologic Targets for Wound Healing and Fibrotic Disorders
by Michael L. Samulevich, Liam E. Carman and Brian J. Aneskievich
Biomedicines 2024, 12(12), 2723; https://doi.org/10.3390/biomedicines12122723 - 28 Nov 2024
Viewed by 334
Abstract
Successful skin wound healing is dependent on an interplay between epidermal keratinocytes and dermal fibroblasts as they react to local extracellular factors (DAMPs, PAMPs, cytokines, etc.) surveyed from that environment by numerous membrane receptors (e.g., TLRs, cytokine receptors, etc.). In turn, those receptors [...] Read more.
Successful skin wound healing is dependent on an interplay between epidermal keratinocytes and dermal fibroblasts as they react to local extracellular factors (DAMPs, PAMPs, cytokines, etc.) surveyed from that environment by numerous membrane receptors (e.g., TLRs, cytokine receptors, etc.). In turn, those receptors are the start of a cytoplasmic signaling pathway where balance is key to effective healing and, as needed, cell and matrix regeneration. When directed through NF-κB, these signaling routes lead to transient responses to the benefit of initiating immune cell recruitment, cell replication, local chemokine and cytokine production, and matrix protein synthesis. The converse can also occur, where ongoing canonical NF-κB activation leads to chronic, hyper-responsive states. Here, we assess three key players, TAK1, TNFAIP3, and TNIP1, in cytoplasmic regulation of NF-κB activation, which, because of their distinctive and yet inter-related functions, either promote or limit that activation. Their balanced function is integral to successful wound healing, given their significant control over the expression of inflammation-, fibrosis-, and matrix remodeling-associated genes. Intriguingly, these three proteins have also been emphasized in dysregulated NF-κB signaling central to systemic sclerosis (SSc). Notably, diffuse SSc shares some tissue features similar to an excessive inflammatory/fibrotic wound response without eventual resolution. Taking a cue from certain instances of aberrant wound healing and SSc having some shared aspects, e.g., chronic inflammation and fibrosis, this review looks for the first time, to our knowledge, at what those pathologies might have in common regarding the cytoplasmic progression of NF-κB-mediated signaling. Additionally, while TAK1, TNFAIP3, and TNIP1 are often investigated and reported on individually, we propose them here as three proteins whose consequences of function are very highly interconnected at the signaling focus of NF-κB. We thus highlight the emerging promise for the eventual clinical benefit derived from an improved understanding of these integral signal progression modulators. Depending on the protein, its indirect or direct pharmacological regulation has been reported. Current findings support further intensive studies of these points in NF-κB regulation both for their basic function in healthy cells as well as with the goal of targeting them for translational benefit in multiple cutaneous wound healing situations, whether stemming from acute injury or a dysregulated inflammatory/fibrotic response. Full article
(This article belongs to the Special Issue Skin Fibrosis and Cutaneous Wound Healing)
Show Figures

Figure 1

Figure 1
<p>TAK1, TNFAIP3, and TNIP1 are focal point for regulating activation of NF-κB, leading to wound healing and fibrotic disorder gene expression (see text). In brief, function of TAK1 leads to progression of cytoplasmic signaling downstream of the indicated receptors, while TNFAIP3 and TNIP1 restrict continuation of the signal. Created in Biorender.com.</p>
Full article ">
15 pages, 4354 KiB  
Article
The Acid-Buffered Engineered Gel Promotes In Vitro Cutaneous Healing and Fights Resistant Bacteria in Wounds
by Fatima Abid, Emmeline Virgo, Tahlia Louise Kennewell, Riya Khetan, Hanif Haidari, Zlatko Kopecki, Yunmei Song and Sanjay Garg
Pharmaceutics 2024, 16(11), 1484; https://doi.org/10.3390/pharmaceutics16111484 - 20 Nov 2024
Viewed by 364
Abstract
Background: Treatment of cutaneous wound infections is becoming a major clinical challenge due to the growing problem of antimicrobial resistance associated with existing wound treatments. Two prevalent pathogens in wound infections, Staphylococcus aureus (S. aureus) and Pseudomonas aeruginosa (P. [...] Read more.
Background: Treatment of cutaneous wound infections is becoming a major clinical challenge due to the growing problem of antimicrobial resistance associated with existing wound treatments. Two prevalent pathogens in wound infections, Staphylococcus aureus (S. aureus) and Pseudomonas aeruginosa (P. aeruginosa), continue to present a serious challenge, underscoring the critical need for new therapeutic alternatives. Methods: Novel alginate acid-buffered gels (ABF-1, ABF-2, and ABF-3) were developed using a combination of organic acids in various concentrations and buffered at a pH of 4.5. The acid-buffering capacity of the gels was evaluated against sodium hydroxide solution and simulated wound fluid (SWF) at different wound pHs, mimicking infected and non-infected wound environments. The in vitro antibacterial activity was assessed against resistant bacterial strains (Gram-positive and Gram-negative) using a microdilution method and wound biofilm assay. The rheological properties and cell viability of the gels were evaluated and the gel showing positive cell viability was further investigated for healing ability using an in vitro wound scratch assay. Results: The gels showed promising in vitro antibacterial activity against Staphylococcus epidermidis, S. aureus, and P. aeruginosa. Gels with higher acid concentrations (ABF-1 and ABF-2) were highly effective in reducing the bacterial load in chronic biofilms of S. aureus and P. aeruginosa, while the gel with a lower acid concentration (ABF-3) showed positive effects on the viability of skin cells (over 80% cells viable) and for promoting wound closure. All three gels demonstrated excellent acid-buffering capabilities. Conclusions: The acid-buffered gels demonstrate promising in vitro antibacterial effects, indicating their potential for enhancing wound healing. Full article
(This article belongs to the Special Issue Prospects of Hydrogels in Wound Healing)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Schematic illustration of the determination of the acid-buffering capacity of gels using SWF (Created in BioRender.com).</p>
Full article ">Figure 2
<p>The buffering activity of ABF-1, ABF-2, and ABF-3 against 1N NaOH solution. The results are presented as mean ± standard deviation, <span class="html-italic">n</span> = 3.</p>
Full article ">Figure 3
<p>The buffering activity of (<b>a</b>) ABF-1, (<b>b</b>) ABF-2, and (<b>c</b>) ABF-3 against SWF at different pH ranges where ‘before’ indicates the pH of the formulation before the start of titration. The results are presented as mean ± standard deviation, <span class="html-italic">n</span> = 3. * shows a significant difference compared to control (<span class="html-italic">p</span> &lt; 0.05) using two-way ANOVA, where ns = non-significant, *** = <span class="html-italic">p</span> &lt; 0.001 and **** = <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 4
<p>Rheological properties of acid-buffered gels measured by shear rate after the equilibrium state at 25 °C.</p>
Full article ">Figure 5
<p>Wound biofilm assay (<b>a</b>) <span class="html-italic">P. aeruginosa</span> and (<b>b</b>) <span class="html-italic">S. aureus</span>. The results are displayed as mean ± standard deviation, <span class="html-italic">n</span> = 3. ** = <span class="html-italic">p</span> &lt; 0.01, *** = <span class="html-italic">p</span> &lt; 0.001, and **** = <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 6
<p>In vitro cell viability analysis in response to treatment with acid-buffered gels in healthy fibroblast and keratinocyte cells. The results are expressed as mean ± standard deviation (<span class="html-italic">n</span> = 3). * Shows a significant difference compared to control (<span class="html-italic">p</span> &lt; 0.05) using two-way ANOVA. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 7
<p>Measurement of wound closure (measured as wound width) and representative images demonstrating the migration of (<b>a</b>) HaCaT and (<b>b</b>) HFF cell lines after the treatment with ABF-3. The error bar represents the mean ± standard deviation (<span class="html-italic">n</span> = 3).</p>
Full article ">
34 pages, 21673 KiB  
Review
Paying Homage to Microvessel Remodeling and Small Vessel Disease in Neurodegeneration: Implications for the Development of Late-Onset Alzheimer’s Disease
by Melvin R. Hayden
J. Vasc. Dis. 2024, 3(4), 419-452; https://doi.org/10.3390/jvd3040033 - 20 Nov 2024
Viewed by 760
Abstract
The microvessel neurovascular unit, with its brain endothelial cells (BEC) and blood–brain barrier remodeling, is important in the development of impaired cognition in sporadic or late-onset Alzheimer’s disease (LOAD), which is associated with aging and is highly prevalent in older populations (≥65 years [...] Read more.
The microvessel neurovascular unit, with its brain endothelial cells (BEC) and blood–brain barrier remodeling, is important in the development of impaired cognition in sporadic or late-onset Alzheimer’s disease (LOAD), which is associated with aging and is highly prevalent in older populations (≥65 years of age). It is also linked with vascular dementia and vascular contributions to cognitive impairment and dementia, including cerebral amyloid angiopathy in neurodegeneration. LOAD is considered to be the number one cause of dementia globally; however, when one considers the role of mixed dementia (MD)—the combination of both the amyloid cascade hypothesis and the vascular hypothesis of LOAD—it becomes apparent that MD is the number one cause. Microvessel BECs are the first cells in the brain to be exposed to peripheral neurotoxins from the systemic circulation and are therefore the brain cells at the highest risk for early and chronic injury. Therefore, these cells are the first to undergo injury, followed by excessive and recurrent wound healing and remodeling processes in aging and other age-related diseases such as cerebrocardiovascular disease, hypertension, type 2 diabetes mellitus, and Parkinson’s disease. This narrative review explores the intricate relationship between microvessel remodeling, cerebral small vessel disease (SVD), and neurodegeneration in LOAD. It also discusses the current understanding of how microvessel dysfunction, disruption, and pathology contribute to the pathogenesis of LOAD and highlights potential avenues for therapeutic intervention. Full article
(This article belongs to the Section Neurovascular Diseases)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Representative transition electron microscopy (TEM) cross-section images of microvessels from various animal models from layer III of the frontal cortex at various magnifications. These images contrast with those of macrovessels that with a diameter measuring ≥5 μm and more than two layers of vascular smooth muscle cell(s) (VSMCs) within their media. Blue open arrows denote the basement membrane of the neurovascular unit in the true capillary. Magnification at 3 μm, 0.5 μm, 5 μm (far-left, middle, and far-right, respectively). Images provided with permission by CC 4.0 [<a href="#B1-jvd-03-00033" class="html-bibr">1</a>,<a href="#B2-jvd-03-00033" class="html-bibr">2</a>]. AC, astrocytes (pseudo-colored gold and blue in far-left and far-right images, respectively). AQP-4, aquaporin 4; AC, perivascular astrocyte; AC1, AC2, astrocyte endfeet numbers 1 and 2; ACef, perivascular astrocyte endfeet; CL, capillary lumen; EC, brain endothelial cell; gs, glymphatic space; lys, lysosome; Mt, mitochondria; N, nucleus; NVU, neurovascular unit; Pc, pericyte; PcN, pericyte nucleus; Pcp, pericyte endfeet processes; PVS, perivascular space; rMGC, interrogating or reactive microglia; rMΦ, reactive macrophage; TJ/AJ, tight junctions/adherens junctions.</p>
Full article ">Figure 2
<p>Perivascular astrocyte endfeet (pvACef) provide the connections between the capillary and regional neurons to form the neurovascular unit (NVU). This combined cartoon illustration and transmission electron micrographs allows one to better visualize how the pvACef connect the NVU capillary to the regional neurons (<b>A</b>). Note that in panel (<b>A</b>) there is detachment and retraction of pvACef, which are noted in the diabetic <span class="html-italic">db/db</span> models. Other pvACef connect with the synapses (<b>B</b>). Modified images in panel (<b>A</b>,<b>B</b>) were provided with permission by CC 4.0 [<a href="#B16-jvd-03-00033" class="html-bibr">16</a>]. Asterisk, denotes emphasis; AC, astrocyte; ACef, astrocyte endfeet; AQP-4, aquaporin four; BDGF, brain derived growth factor; Ca++, calcium ion; CKC, control model; DBC, diabetic <span class="html-italic">db/db</span> model; DBE, diabetic <span class="html-italic">db/db</span> treated with empagliflozin; GDGF, glioma-derived growth factor; PSD, post synaptic density; S, synapse; TGF-β, transforming growth factor beta.</p>
Full article ">Figure 3
<p>Multiple peripheral-systemic injurious species (neurotoxins) affect the brain endothelial cells (BECs) of the brain. These injurious species activate the BECs of the neurovascular unit (NVU), resulting in BEC activation and dysfunction (BEC<span class="html-italic">act/dys</span>) and blood–brain barrier dysfunction/disruption (BBB<b><span class="html-italic">dd</span></b>). BEC<span class="html-italic">act/dys</span> and BBB<b><span class="html-italic">dd</span></b> are biomarkers for the development of cerebral small vessel disease (SVD). Note the red-dashed line at the top of this image, which designates the location of the multiple injurious species that are responsible for initial brain endothelial cell injury in multiple clinical diseases and structural abnormalities, including SVD. BEC insulin resistance associated with an increase in glucose and the AGE receptor (RAGE) interaction is also important in the development of BECact/dys, BBB<b><span class="html-italic">dd</span></b>, and neurodegeneration (not shown). Image provided with permission by CC 4.0 [<a href="#B36-jvd-03-00033" class="html-bibr">36</a>,<a href="#B39-jvd-03-00033" class="html-bibr">39</a>]. AGE, advanced glycation end products; Ang II, angiotensin two; BBB, blood–brain barrier; BEC, brain endothelial cell; BBB<b><span class="html-italic">dd</span></b>, blood–brain barrier dysfunction and disruption; BECact/dys, brain endothelial cell activation/dysfunction; BH4, tetrahydrobiopterin; CCL2, chemokine (C-C motif) ligand 2; Cox-2, cyclo-oxygenase-2; Cox-2/PGE2 axis, cyclo-oxygenase-2/prostaglandin E2; ecGCx, endothelial glycocalyx; ICAM-1, intercellular adhesion molecule-1; IL-1β, interleukin-1β; IL-6, interleukin-6; JAMs, junctional adhesion molecules; LDL, low-density lipoprotein cholesterol; LPa, lipoprotein little a; MCP-1, monocyte chemotactic protein-1; NO, nitric oxide; Nox2, (NADPH Ox (nicotinamide adenine dinucleotide phosphate oxidase); ONOO-, peroxinitrite; pnsCC, peripheral nervous system cytokines and chemokines; NVU, neurovascular unit; RBC, red blood cell; Red arrows, denote increase; RONSS, reactive oxygen, nitrogen, sulfur species; ROS, reactive oxygen species; RSI, reactive species interactome; T, transcytosis; TNFα, tumor necrosis factor alpha; VCAM-1, vascular cellular adhesion molecule-1; WBC, white blood cell.</p>
Full article ">Figure 4
<p>Brain endothelial cell activation and dysfunction (BEC<span class="html-italic">act/dys</span>) phenotypes in obese 20-week-old female diabetic <span class="html-italic">db/db</span> models. (<b>A</b>,<b>B</b>) demonstrate normal control phenotypes of microvessel neurovascular unit (NVU) BEC from control C57B5J model at 20 weeks in the frontal cortical layer III in cross and longitudinal sections (<b>A</b>,<b>B</b>), respectively. Note that the perivascular astrocytes are pseudo-colored golden with apparently normal electron-dense mitochondria. Also note the cyan-colored line that demarcates the glia limitans in (<b>A</b>,<b>B</b>). (<b>C</b>) demonstrates the control normal phenotype in the C57B6J model at 20 weeks. (<b>D</b>) depicts an activated BEC with marked abrupt swelling of the BEC that is hyperlucent as compared to the adjacent normal thickness of the BEC, which depicts BECact/dys phenotype in the 20-week-old db/db model from the frontal cortex of layer III. (<b>E</b>) depicts the BM thickening of BECs in (<b>D</b>). Note the vacuole-like structures (V) within the BM. (<b>F</b>) through (I) depict the adhesion of monocyte (<b>F</b>), a lymphocyte (<b>G</b>), a platelet (<b>H</b>), and a red blood cell (RBC) (<b>I</b>). Modified images provided with permission by CC 4.0 [<a href="#B2-jvd-03-00033" class="html-bibr">2</a>,<a href="#B39-jvd-03-00033" class="html-bibr">39</a>,<a href="#B40-jvd-03-00033" class="html-bibr">40</a>,<a href="#B41-jvd-03-00033" class="html-bibr">41</a>]. AC, astrocyte; ACfp, astrocyte foot process endfeet; BM, basement membrane; Cap L, capillary lumen; CL, capillary lumen; ECact, brain endothelial cell activation; iMGC, interrogating microglia cell; Mt, mitochondria; Mp, microparticles; Pc, pericyte; Red arrows, denote BEC activation; White arrows, denote RBC adhesion plaques; Yellow arrows, denote platelet.</p>
Full article ">Figure 5
<p>Proinflammatory LPS results in attenuation and discontinuous endothelial glycocalyx (ecGCx) and increased pinocytosis/transcytosis in activated brain endothelial cell activation/dysfunction (BEC<span class="html-italic">act/dys</span>). Far-<b>left</b> panels (<b>B</b>,<b>D</b>) depict an attenuation and discontinuous ecGCx with large gaps of BECs as compared to controls with an intact and continuous ecGCx, as in panels (<b>A</b>,<b>C</b>). Far-<b>right</b> panels (<b>B</b>,<b>D</b>) depict increased pinocytosis/transcytosis of BEC as compared to control panels (<b>A</b>,<b>C</b>). Panels (<b>C</b>,<b>D</b>) are illustrations to improve and highlight the findings of the TEMs depicted in panels (<b>A</b>,<b>B</b>). Images provided with permission by CC 4.0 [<a href="#B42-jvd-03-00033" class="html-bibr">42</a>]. Asterisks, denote macropinocytosis; ACfp, astrocyte foot process endfeet; atMGC, attracted microglia cell(s); BECact/dys, brain endothelial cell activation/dysfunction; CL, capillary lumen; ecGCx, brain endothelial cell glycocalyx; EC N, brain endothelial cell nucleus; LPS, lipopolysaccharide; Pc N, pericyte nucleus; PVS, perivascular space; TJ/AJ, tight and adherence junctions.</p>
Full article ">Figure 6
<p>Loss and/or disruption of the normal continuous tight and adherence junction(s) (TJ/AJs) paracellular blood–brain barrier (BBB) in male CD-1 streptozotocin-induced (STZ) diabetic preclinical mice models, resulting in blood–brain barrier dysfunction and disruption (BBB<b><span class="html-italic">dd</span></b>) protected by the carbonic anhydrase inhibitor (topiramate, a mitochondria-specific antioxidant) in the midbrain as compared to the cerebellum. STZ-induced diabetic mice revealed disruption of the BBB by <sup>14</sup>C-sucrose measurements. Panel (<b>A</b>) displays three prominent elongated and continuous highly electron-dense TJ/AJ (white and black arrows). Panel (<b>B</b>) depicts a discontinuous and disrupted TJ/AJ (black arrows) into three distinct segments in the midbrain of STZ-induced diabetic models. Note how TJ/AJs tend to form at the BEC-BEC overlap junctions in panels (<b>A</b>–<b>C</b>). Panel (<b>C</b>) illustrates that treatment with topiramate (TOP) prevented disruption in the brain endothelial cell BBB (<span class="html-italic">yellow</span> and black arrows and yellow dashed line below the intact BBB TJ/AJ) in the midbrain. Revised figure images provided with permission by CC 4.0 [<a href="#B39-jvd-03-00033" class="html-bibr">39</a>,<a href="#B42-jvd-03-00033" class="html-bibr">42</a>,<a href="#B43-jvd-03-00033" class="html-bibr">43</a>]. Magnification ×3000; scale bar = 0.5 μm (<b>A</b>); ×10,000; scale bar = 0.2 μm in (<b>B</b>,<b>C</b>). BEC, brain endothelial cell; EC, brain endothelial cell; CL, capillary lumen; Pc, pericyte; RBC, red blood cell; Rx, treatment; T1DM, type 1 diabetes mellitus; TOP, topiramate; White arrows, denote brain endothelial cell.</p>
Full article ">Figure 7
<p>Brain pericyte(s) (Pcs) and brain endothelial cell(s) (BECs) are closely interconnected and are interdependent for paracrine signaling, making the Pc a critical cell for proper homeostatic function of the neurovascular unit(s) (NVU) in controlling regional cerebral blood flow (CBF). Panel (<b>A</b>) demonstrates the normal arrangement of the BEC, Pc, and perivascular astrocyte endfeet (pvACef). Note how the basement membrane (black open arrows) is interspersed and separates the luminal BEC, Pc, and the more abluminal pvACef. Also note the interrogating microglia cell (iMGC) (white closed arrow). Panel (<b>B</b>) illustrates with better clarity the close interactions between the Pc and the BEC (EC), which importantly allows for paracrine signaling as well as structural and functional support by the NVUs. Panel (<b>C</b>) illustrates the paracrine signaling of nitric oxide (NO) synthesized by the eNOS enzyme to signal the Pc cell to relax, allowing for vasodilation and the supportive connection by the platelet-derived growth factor beta (PDGFβ) of the EC to interact with the PDGFβ receptor (PDGFβ R) of the adjacent Pc. Images provided with permission by CC 4.0 [<a href="#B44-jvd-03-00033" class="html-bibr">44</a>,<a href="#B45-jvd-03-00033" class="html-bibr">45</a>]. ACef, astrocyte endfeet; AQP4, aquaporin 4; PcP, pericyte processes-endfeet-foot processes; N, nucleus; TJ/AJ, tight and adherens junctions; VEGF A-B, vascular endothelial growth factor A-B; VEGF R, receptor for VEGF.</p>
Full article ">Figure 8
<p>Pericyte endfeet (Pcef) retraction and pericyte nucleus (Pc N) rounding are early signs of Pc dysfunction, along with the formation of aberrant mitochondria (aMt), which are found in brain endothelial cells (ECs), Pcs, astrocyte(s) (ACs), neuronal axons, and oligodendrocytes (OLIG) along with Pc cytoplasmic apoptotic changes with apoptotic bodies. Panels (<b>1</b>–<b>3</b>) illustrate Pc N rounding and retraction of Pcef in panels (<b>2</b> and <b>3</b>) as compared to control model Pc N elongation and elongation of Pcef (pseudo-colored green) in panel 1. Panels (<b>A</b>–<b>E</b>) depict aberrant mitochondria (aMt) in BECs, ACs, Pcef, neuronal axons, and oligodendrocytes (OLIG), respectively. Panel (<b>F</b>) depicts an apoptotic Pc with numerous apoptotic bodies observed in this Pc cytoplasm (arrows), which are indicative of Pc degeneration and death. Scale bars vary and are included in the images presented. Modified images provided with permission by CC 4.0 [<a href="#B59-jvd-03-00033" class="html-bibr">59</a>,<a href="#B60-jvd-03-00033" class="html-bibr">60</a>,<a href="#B61-jvd-03-00033" class="html-bibr">61</a>]. Asterisk, denotes emphasis; BEC, brain endothelial cell; CL, capillary lumen; EC, endothelial cell-brain endothelial cell; N, nucleus; Pc, pericyte; RBC, red blood cell.</p>
Full article ">Figure 9
<p>Neurotoxicity of amyloid beta in close proximity to the neurovascular unit (NVU) brain endothelial cell(s) (BECs) may result in blood–brain barrier dysfunction and disruption (BBB<b><span class="html-italic">dd</span></b>). These images are from a 9-month-old 5xFAD male mouse model not previously published. Panel (<b>A</b>) depicts interstitial extracellular matrix-interstitial amyloid beta (Aβ) (pseudo-colored red) in close proximity to the NVU, which appears to nearly touch the outer basement membrane (BM) of the NVU depicted. Importantly, note that this image depicts degenerative neurites (yellow arrows and outlined in yellow dashed-lines) within the adjacent interstitial neuropil. Panel (<b>B</b>) depicts an exploded image in Microsoft paint with an intact scale bar. Insert (<b>C</b>) is a further exploded image demonstrating how Aβ is closely adjacent to the microvessel NVU BEC basement membrane (BM) (open red and white arrows) and appears to actually be in direct contact with the BM. Indeed, this structural arrangement contributes to Aβ being neurotoxic to BECs and creating a vicious cycle of BEC injury by Aβ and BECact/dys that contributes to a further increase in ROS and damage to the NVU, with eventual NVU uncoupling and a decrease in regional cerebral blood flow. This will ultimately increase neurodegeneration and increase or accelerate amyloid beta deposition.</p>
Full article ">Figure 10
<p>Astrocyte(s) (ACs) are the master communication/connecting cell(s) (CCC) within the brain universe. This collection of illustrations and transmission electron microscopy (TEM) images demonstrates the CCC functions of ACs through their various perivascular, perisynaptic, perineuronal endfeet, and cell-cell junctions (inserts 1–5). Insert 1 demonstrates the important role of perivascular astrocyte endfeet (pvACef; pseudo-colored golden) communication/connection. Note how this communicating/connecting AC allows for neurovascular coupling with regional neurons (insert 3) in frontal cortex layer III in control mice at 20 weeks of age. Insert 2 illustrates the communication/connection of the astrocyte perisynaptic endfeet (psACef) (pseudo-colored yellow). Insert 3 illustrates the communication/connection of ACs to myelinated and unmyelinated neurons. Insert 4 depicts the lost connections between a reactive microglial cell (<span class="html-italic">r</span>MGC) (pseudo-colored blue with nuclear chromatin condensation) and multiple reactive, detached, and separated ACs (pseudo-colored red) adjacent to a neurovascular unit (NVU) with a single intact non-reactive AC (pseudo-colored yellow) in the diabetic <span class="html-italic">db/db</span> model cortical layer III at 20 weeks of age. Insert 5 illustrates AC-to-AC connections in cortical layer III in control models (hand-drawn computer-assisted illustration of light microscopic toluidine blue stained images from control C57BL/6J models) via gap junction connexins (Cx43). Inserts 1–4 have scale bars of 0.5 μm, 100 nm, 1 μm, and 2 μm, respectively. The background image represents a hand-drawn computer-assisted image derived from control C57BL/6J toluidine blue stained models and does not have a scale bar. This highly modified image is provided with permission by CC 4.0 [<a href="#B16-jvd-03-00033" class="html-bibr">16</a>]. ACfp, protoplasmic perivascular astrocyte endfeet; ACPVef, astrocyte perivascular endfeet; Cap L, capillary lumen; EC, brain endothelial cell; iMGC, interrogating microglial cell; Mt, mitochondria; N, nucleus; Pc, pericyte; PSD, post-synaptic density; PVACef, perivascular astrocyte endfeet; rMGC, reactive microglia cell; psACef, perisynaptic astrocyte endfeet.</p>
Full article ">Figure 11
<p>A master communicating, connecting, and signaling cell of the brain universe providing homeostasis. In addition to being neuroprotective, astrocytes provide molecular, cellular, and network communication, as well as systemic, metabolic, and whole organ homeostasis. The centrally located modified image of an astrocyte from <a href="#jvd-03-00033-f010" class="html-fig">Figure 10</a> was provided with permission by CC 4.0 [<a href="#B16-jvd-03-00033" class="html-bibr">16</a>]. AC, astrocyte; ACef, astrocyte endfeet; AQP4, aquaporin-4; BBB, blood–brain barrier; Ca++, calcium; Cap L, capillary lumen; CBF, cerebral blood flow; Cl-, chloride; CNS, central nervous system; CO<sub>2</sub>, carbon monoxide; EC, brain endothelial cell; GABA, gamma aminobutyric acid; K+, potassium; Na+, sodium; NVU, neurovascular unit; PVACef, perivascular astrocyte endfeet; rMGC, reactive microglia cell.</p>
Full article ">Figure 12
<p>Reactive microglia cell(s) (rMGCs) and astrocyte(s) (rACs) contribute to Ddecreased cerebral blood flow and neurodegeneration (ND). Panel (<b>A</b>) demonstrates a normal neurovascular unit (NVU) with its normal interrogating MGC (iMGC) (pseudo-colored green) and its intact astrocytes (iAC) (pseudo-colored golden). Panel (<b>B</b>) depicts a prominent rMGC invading the NVU (pseudo-colored red). Note the aberrant mitochondria (aMt) (pseudo-colored yellow and outlined in red). Notably, the normal iAC have become detached and retracted, separated (drAC, pseudo-colored light blue) from the NVU outer basement membrane and results in NVU uncoupling, with ensuing decreased cerebral blood flow to regional neurons resulting in hypometabolism, hypoperfusion, regional neuronal hypoxia/ischemia with subsequent neurodegeneration, and impaired cognition. Modified images provided with permission by CC 4.0 [<a href="#B6-jvd-03-00033" class="html-bibr">6</a>,<a href="#B60-jvd-03-00033" class="html-bibr">60</a>]. CL, capillary lumen; drAC, detached reactive, retracted astrocyte endfeet; N, nucleus; RBC, red blood cell.</p>
Full article ">Figure 13
<p>Enlarged perivascular space (EPVS) and resident-reactive perivascular macrophage (rPVMΦs) in a postcapillary venule compared to a true capillary. Panel (<b>A</b>) demonstrates a normal true capillary in a 20-week-old female C57B6/J control model. Note how the astrocyte endfeet (ACef) tightly abut the shared basement membrane (open black arrows) of the brain endothelial cell (BEC) and pericyte foot process (PcP). Panel (<b>B</b>) depicts an EPVS with a prominent rPVMΦ (pseudo-colored red) in a 20-week-old lipopolysaccharide (LPS)-treated CD-1 male mouse model. Note how the ACfp are markedly separated from the capillary mural cells (BEC and Pc) (red double arrows). Panel (<b>C</b>) depicts the rPVMΦs in an exploded image with intimate contact with the Pcfps basal lamina, as well as its intimate contact with the basal lamina of the ACef (outermost boundary of the EPVS abluminal lining) (dashed blue circles). Modified images provided with permission by CC 4.0 [<a href="#B45-jvd-03-00033" class="html-bibr">45</a>]. AQP4, aquaporin 4; Lys, lysosomes; Mt, mitochondria; N, nucleus; NVU, neurovascular unit; V, vacuoles; ves, vesicles.</p>
Full article ">Figure 14
<p>Multipotential reactive astrocyte (rAC) phenotype remodeling in late-onset Alzheimer’s disease (LOAD) from hypertrophy to atrophy. Panel (<b>A</b>) depicts extracellular matrix interstitial space amyloid beta (Aβ) plaques that are increased in LOAD and co-occur with soluble Aβ oligomers, which are actually more neurotoxic than the Aβ mature plaques. Panel (<b>B</b>) lists the components of Aβ plaques (1.-5.). Panel (<b>CI</b>) demonstrates the remodeling of the rACs that follows such injuries as ischemic or hemorrhagic stroke with palisading and hypertrophic phenotypes in contrast to the rAC phenotype of atrophy, which is associated with AC remodeling due to the neurotoxicity of Aβ oligomers and Aβ extracellular plaque aggregation, as discussed in (<b>CII</b>). Modified images of Aβ in panel (<b>A</b>) are provided with permission by CC 4.0 [<a href="#B74-jvd-03-00033" class="html-bibr">74</a>] and the reproduced image in C1 is provided with permission by CC 4.0 [<a href="#B28-jvd-03-00033" class="html-bibr">28</a>,<a href="#B75-jvd-03-00033" class="html-bibr">75</a>]. ApoE, apolipoprotein E; CNS, central nervous system; cnsCC, central nervous system cytokines/chemokines; DNA, deoxyribonucleic acid; GAGS, glycosaminoglycans; LOAD, late-onset Alzheimer’s disease; MMP(s), matrix metalloproteinases; PGN, proteoglycan(s); rAC(s), reactive astrocytes; ROI, region of interest; rMGC(s), reactive microglia cells; ROS, reactive oxygen species; RNA, ribonucleic acid.</p>
Full article ">Figure 15
<p>Aberrant mitochondria (aMt) allow for leaky prooxidative ROS of the reactive species interactome (RSI) and prooxidative iron sulfur cluster(s) (ISCs) to enter the cytosol with dysfunction and damage to the cell and surrounding cells. Note how the aMt remodeling appears as compared to the normal control insert upper left with its hyperlucency, crista fragmentation and loss, loss of electron dense mitochondrial matrix, and permeabilization of the outer mitochondria membrane (asterisk). This permeabilization allows the redox-active and prooxidative mtROS and iron sulfur clusters to escape into the cellular cytosol resulting in cellular dysfunction, damage and even apoptosis. This prooxidant mechanism has an increased impact if the antioxidant reserves of super oxide dismutase (SOD), catalase, and glutathione (GSH) have previously been or are currently depleted. Revised background image of the aMt is provided with permission by CC 4.0 [<a href="#B61-jvd-03-00033" class="html-bibr">61</a>]. aMt, aberrant mitochondria; Cys, cysteine; ETC, electron transport chain; Fe, iron; ISC, iron sulfur cluster(s); MOMP, mitochondrial outer membrane permeabilization; Mt, mitochondria; mtROS, mitochondrial-derived reactive oxygen species; RONSS, reactive oxygen, nitrogen, sulfur species; S, sulfur.</p>
Full article ">Figure 16
<p>Reactive oxygen species (ROS) instigates ROS and oxidative-redox stress (OxRS) in a vicious cycle with neuroinflammation. This vicious cycle contributes to impaired cognition and neurodegeneration. Metabolic and hormonal excesses in addition to injury-trauma and the vicious cycle of neuroinflammation and ROS—OxRS lead to blood–brain barrier dysfunction and disruption with NVU uncoupling and regional hypometabolism and ischemia/ischemia reperfusion injury result in a cascade to neurodegeneration and brain atrophy with impaired cognition that associates with late-onset Alzheimer’s disease (LOAD). Modified figure provided with permission by CC 4.0 [<a href="#B28-jvd-03-00033" class="html-bibr">28</a>,<a href="#B36-jvd-03-00033" class="html-bibr">36</a>,<a href="#B85-jvd-03-00033" class="html-bibr">85</a>]. AGE/RAGE, advanced glycation end products/receptor for advanced glycation end products; Ang II, angiotensin II; AT1R, angiotensin type 1 receptor; BH4, tetrahydrobiopterin; eNOS, endothelial nitric oxide synthase; FFA, free fatty acids; HPA, hypothalamic pituitary adrenal; I-CAM, intercellular adhesion molecule; MC, mast cell; MGCs, microglia cells; NADPH—NADPH Ox, reduced nicotinamide adenine dinucleotide phosphate oxidase; NVU. neurovascular unit; NF-kB, nuclear factor- kappa B; RAS, renin angiotensin system; RAAS, renin angiotensin aldosterone system; ROS/RNS, reactive oxygen species/reactive nitrogen species. UV, ultraviolet; VE-CAM, vascular endothelial cellular adhesion molecule.</p>
Full article ">Figure 17
<p>Vicious cycles and multiple injurious hits combine to result in small vessel disease (SVD), neuronal injury, neurodegeneration, brain atrophy, impaired cognition, and LOAD. Panel (<b>A</b>) depicts the importance of the vicious cycle between oxidative-redox stress (OxRS) and inflammation resulting in microvessel remodeling and cerebral small vessel disease (SVD), which contribute to impaired cognition and the development of neurodegeneration. Panel (<b>B</b>) depicts the multiple injurious hits that trigger the OxRS and neuroinflammation, which are involved in the development of neuronal injury and neurodegeneration. Panel (<b>A</b>) is reproduced with permission by 4.0 [<a href="#B36-jvd-03-00033" class="html-bibr">36</a>]. Act, activation; BBB, blood–brain barrier; BBB<b><span class="html-italic">dd</span></b>, blood–brain barrier dysfunction and disruption; BEC, brain endothelial cell; CMBs, cerebral microbleeds; cnsCC, central nervous system cytokines/chemokines; dys, dysfunction; EPVS, enlarged perivascular spaces; MMP, matrix metalloproteinases; NFkappaB, nuclear factor kappa beta; <b><span class="html-italic">p</span></b>CC, peripheral cytokines/chemokines; ROS, reactive oxygen species; RONSS, reactive oxygen, nitrogen, sulfur species; RSI, reactive species interactome; TJ/AJ, tight and adherens junctions; VE, vascular endothelial.</p>
Full article ">Figure 18
<p>Neuroinflammation induces neurodegeneration via proinflammatory central nervous system cytokines and chemokines (<b><span class="html-italic">cns</span></b>CC), amyloid beta (Aβ), and tau. (<b>A</b>) reveals a cleaned transmission electron microscopy (TEM) image of a reactive microglia (rMGC). (<b>B</b>) depicts an illustration of a reactive astrocyte (rAC). (<b>C</b>) represents an illustration of a myelinated neuron. (<b>D</b>) depicts a portion of a myelinated neuronal axon that importantly depicts aberrant mitochondria (aMt) capable of leaking mitochondria reactive oxygen species (mtROS) and splitting of myelin that could impair informational transfer. (<b>E</b>) depicts a TEM image of a dendritic neurite (dn) outlined with yellow-dashed lines. (<b>F</b>,<b>G</b>) depict a TEM image of an Aβ plaque outlined by yellow-dashed lines. (<b>H</b>,<b>I</b>) depict immunohistologic light microscope images of Aβ. Image G scale bar = 5 μm. (<b>I</b>) scale bar = 20 μm. Modified (<b>A</b>,<b>D</b>) provided with permission by CC 4.0 [<a href="#B60-jvd-03-00033" class="html-bibr">60</a>,<a href="#B61-jvd-03-00033" class="html-bibr">61</a>]. (<b>H</b>,<b>I</b>) provided with permission by CC 4.0 [<a href="#B74-jvd-03-00033" class="html-bibr">74</a>]. aMt, aberrant mitochondria; CC, chromatin condensation; COX, cyclooxygenase; cnsCC, central nervous system cytokines chemokines; dn, dendritic neurite; IL-1, interleukin 1; IL-1β, interleukin 1 beta; IL-6, interleukin 6; ISC, iron sulfur clusters; Lys, lysosome; mtROS, mitochondria reactive oxygen species; N, nucleus; NADPH Ox, nicotine adenine diphosphate oxidase; NFTs, neuro fibrillary tangles; NO, nitric oxide; NOX, NADPH Ox; OxRS, oxidative redox stress; rACs, reactive astrocytes; rMGCs, reactive microglia cells; TNFα, tumor necrosis alpha.</p>
Full article ">Figure 19
<p>Microvessel SVD definitely contributes to and are linked to the development of neurodegeneration. At least a half dozen mechanisms are involved. Importantly, note that number 5 points to the quartet of mechanisms involved in this linkage of SVD to neurodegeneration. Importantly these six mechanisms of linage seem to be initiated by brain endothelial cell activation and dysfunction (BEC<span class="html-italic">act/dys</span>) with concurrent blood–brain barrier dysfunction and/or disruption (BBBdd). Aβ, amyloid beta; CBF, cerebral blood flow; EPVS, enlarged perivascular spaces, LOAD, late-onset Alzheimer’s disease; SVD, small vessel disease.</p>
Full article ">Figure 20
<p>Gross brain atrophy in the <span class="html-italic">db/db</span> obese diabetic model but not in the obese diabetic BTBR <span class="html-italic">ob/ob</span> model or obese and diabetic <span class="html-italic">db/db</span> models treated with empagliflozin. Panel (<b>A</b>) demonstrates the normal gross brain in the control male model at 20-weeks of age. Panel (<b>B</b>) depicts marked brain atrophy at the time of surgical removal of the diabetic <span class="html-italic">db/db</span> models as compared to control models in Panel (<b>A</b>). Note the marked atrophy or loss in the cortical-parietal-hippocampal regions (outlined by the yellow dashed lines) in panel (<b>B</b>) as compared to the control in panel (<b>A</b>) at 26-weeks of age and these remodeling changes were associated with a decrease in the brain wet weights upon removal. Panel (<b>C</b>) demonstrates the absence of remodeling atrophy in the <span class="html-italic">ob/ob</span> diabetic obese model at 20-weeks of age and also those db/db diabetic models treated with empagliflozin at 26-weeks of age. Modified panels (<b>A</b>,<b>B</b>) were provided with permission by CC 4.0 [<a href="#B114-jvd-03-00033" class="html-bibr">114</a>,<a href="#B115-jvd-03-00033" class="html-bibr">115</a>].</p>
Full article ">Figure 21
<p>Vascular contributions to cognitive impairment and dementia (VCID) are central to the development of neurodegeneration and late-onset Alzheimer’s disease (LOAD) and/or mixed dementia (MD). Cardio-cerebrovascular diseases including stroke (ischemic and hemorrhagic) associate with and contribute to brain endothelial cell activation and dysfunction (BEC<span class="html-italic">act/dys</span>) with concurrent blood–brain barrier dysfunction and disruption (BBB<b><span class="html-italic">dd</span></b>) induced ischemia, hypoxia, and chronic cerebral hypoperfusion (CCH) to result in neurodegeneration and dementia in LOAD and MD.</p>
Full article ">Figure 22
<p>The dementia quartet mechanisms intersect and contribute to neurodegeneration with neuronal synapse and neuron dysfunction and or loss with regional brain atrophy in late-onset Alzheimer’s disease (LOAD) and mixed dementia (MD) in addition to the small vessel disease (SVD) intersection and contribution to neurodegeneration, LOAD, and MD.</p>
Full article ">Figure 23
<p>Future directions in microvessel small vessel disease that contribute to late-onset Alzheimer’s disease. BEC<span class="html-italic">act/dys</span>, brain endothelial cell activation and dysfunction; BBB<b><span class="html-italic">dd</span></b>, blood–brain barrier dysfunction and disruption; BH4, tetrahydrobiopterin; <b><span class="html-italic">cns</span></b>CC, central nervous system cytokines chemokines; eNOS, endothelial nitric oxide synthase; H2S, hydrogen sulfide; LOAD, late-onset Alzheimer’s disease; MMP, matrix metalloproteinase; NO, nitric oxide; OxRS, oxidative redox stress; <b><span class="html-italic">p</span></b>CC, peripheral cytokines chemokines; SVD, small vessel disease; T, tesla; tPA, tissue-type plasminogen activator; VaD/VAD, vascular dementia.</p>
Full article ">
12 pages, 1398 KiB  
Article
Wound Gel Formulations Containing Poloxamer 407 and Polyhexanide Have In Vitro Antimicrobial and Antibiofilm Activity Against Wound-Associated Microbial Pathogens
by Jeyachchandran Visvalingam, Nandadeva Yakandawala, Suresh Regmi, Adetola Adeniji, Parveen Sharma and Miloslav Sailer
Microorganisms 2024, 12(11), 2362; https://doi.org/10.3390/microorganisms12112362 - 19 Nov 2024
Viewed by 723
Abstract
Chronic wounds are often caused or exacerbated by microbial biofilms that are highly resistant to antimicrobial treatments and that prevent healing. This study compared the antimicrobial and antibiofilm activity of nine topical wound treatments, comprising gels with different concentrations of poloxamer 407 (20–26%) [...] Read more.
Chronic wounds are often caused or exacerbated by microbial biofilms that are highly resistant to antimicrobial treatments and that prevent healing. This study compared the antimicrobial and antibiofilm activity of nine topical wound treatments, comprising gels with different concentrations of poloxamer 407 (20–26%) and different pH levels (4–6) and containing polyhexanide (PHMB) as an antimicrobial agent; the effects of pH on wound gels containing this agent have not been previously reported. The wound gel formulations were tested against six common wound-associated microbial pathogens: Staphylococcus aureus, S. epidermidis, Pseudomonas aeruginosa, Escherichia coli, Acinetobacter baumannii, and Candida albicans. Time-kill assays were used to assess antimicrobial activity against planktonic forms of each species, and a colony biofilm model was used to assess antibiofilm activity against existing biofilms as well as inhibition of new biofilm formation. Biofilm inhibition activity was also assessed in the presence of common wound dressing materials. Wound gels with higher pH levels exhibited stronger antimicrobial activity, while poloxamer 407 concentrations >20% negatively impacted antimicrobial activity. Wound gel formulations were identified that had antimicrobial, antibiofilm, and biofilm inhibition activity against all tested species in vitro. Biofilm inhibition activity was not affected by contact with common wound dressings. Further development of these wound gels may provide a valuable new option for the treatment and prevention of chronic wounds. Full article
Show Figures

Figure 1

Figure 1
<p>Antibiofilm activity of wound gel formulations against preformed biofilms of (<b>A</b>) <span class="html-italic">S. aureus</span>, (<b>B</b>) <span class="html-italic">S. epidermidis</span>, (<b>C</b>) <span class="html-italic">A. baumannii</span>, (<b>D</b>) <span class="html-italic">P. aeruginosa</span>, (<b>E</b>) <span class="html-italic">E. coli</span>, and (<b>F</b>) <span class="html-italic">C. albicans</span> after two successive 24 h application of the gel. NT, no treatment. Mean values identified by different alphabets are significantly different (<span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 2
<p>Biofilm inhibition activity of wound gel formulation P1K3 against six microbial species.</p>
Full article ">Figure 3
<p>Biofilm inhibition activity of wound gel formulation P1K3 in the presence of common wound dressing materials. Mean values of each organism identified by different alphabets are significantly different (<span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">
18 pages, 1486 KiB  
Review
Impact of Hyaluronic Acid and Other Re-Epithelializing Agents in Periodontal Regeneration: A Molecular Perspective
by Alessandro Polizzi, Ylenia Leanza, Antonio Belmonte, Cristina Grippaudo, Rosalia Leonardi and Gaetano Isola
Int. J. Mol. Sci. 2024, 25(22), 12347; https://doi.org/10.3390/ijms252212347 - 17 Nov 2024
Viewed by 818
Abstract
This narrative review delves into the molecular mechanisms of hyaluronic acid (HA) and re-epithelializing agents in the context of periodontal regeneration. Periodontitis, characterized by chronic inflammation and the destruction of tooth-supporting tissues, presents a significant challenge in restorative dentistry. Traditional non-surgical therapies (NSPTs) [...] Read more.
This narrative review delves into the molecular mechanisms of hyaluronic acid (HA) and re-epithelializing agents in the context of periodontal regeneration. Periodontitis, characterized by chronic inflammation and the destruction of tooth-supporting tissues, presents a significant challenge in restorative dentistry. Traditional non-surgical therapies (NSPTs) sometimes fail to fully manage subgingival biofilms and could benefit from adjunctive treatments. HA, with its antibacterial, antifungal, anti-inflammatory, angiogenic, and osteoinductive properties, offers promising therapeutic potential. This review synthesizes the current literature on the bioactive effects of HA and re-epithelializing agents, such as growth factors and biomaterials, in promoting cell migration, proliferation, and extracellular matrix (ECM) synthesis. By modulating signaling pathways like the Wnt/β-catenin, TGF-β, and CD44 interaction pathways, HA enhances wound healing processes and tissue regeneration. Additionally, the role of HA in facilitating cellular crosstalk between epithelial and connective tissues is highlighted, as it impacts the inflammatory response and ECM remodeling. This review also explores the combined use of HA with growth factors and cytokines in wound healing, revealing how these agents interact synergistically to optimize periodontal regeneration. Future perspectives emphasize the need for further clinical trials to evaluate the long-term outcomes of these therapies and their potential integration into periodontal treatment paradigms. Full article
(This article belongs to the Special Issue Periodontitis: Advances in Mechanisms, Treatment and Prevention)
Show Figures

Figure 1

Figure 1
<p>Structure of disaccharide repeating unit of HA. The unit of HA is composed of β-(1,4)-glucuronic acid and β-(1,3)-N-acetylglucosamine linked together by β-1,3 and β-1,4 glycosidic bonds. The molecular weight of this molecule depends on the number of repetitions of the disaccharide unit (n).</p>
Full article ">Figure 2
<p>Wound healing process. It comprises hemostasis, inflammation, proliferation, and remodeling. Reprinted with permission from [<a href="#B20-ijms-25-12347" class="html-bibr">20</a>]. Copyright 2024 American Chemical Society.</p>
Full article ">Figure 3
<p>Schematic representation of periodontal tissue engineering.</p>
Full article ">Figure 4
<p>The synthesis and effects of PGE2 in periodontitis. From [<a href="#B72-ijms-25-12347" class="html-bibr">72</a>], under Creative Commons Attribution 4.0 International License.</p>
Full article ">
14 pages, 4959 KiB  
Article
Enhancing Skin Wound Healing in Diabetic Mice Using SIKVAV-Modified Chitosan Hydrogels
by Xionglin Chen, Xiaoming Cao, Jie Zhang, Chen Jiang, Yitian Yu and Hui Chen
Molecules 2024, 29(22), 5374; https://doi.org/10.3390/molecules29225374 - 14 Nov 2024
Viewed by 424
Abstract
Diabetic foot ulcers (DFUs), a prevalent chronic disease caused by various factors, significantly impact patients’ quality of life due to prolonged healing times and increased infection risks. Current treatment modalities, including pharmacological, physical, and surgical interventions, often yield limited efficacy and adverse effects, [...] Read more.
Diabetic foot ulcers (DFUs), a prevalent chronic disease caused by various factors, significantly impact patients’ quality of life due to prolonged healing times and increased infection risks. Current treatment modalities, including pharmacological, physical, and surgical interventions, often yield limited efficacy and adverse effects, highlighting the urgent need for novel therapeutic strategies. The objective of this research is to create SIKVAV-modified chitosan hydrogels with the intention of improving the process of skin wound healing in diabetic mice. We synthesized the hydrogels and established a diabetic mice model with skin wounding to evaluate its healing effects and underlying mechanisms. The results of our study indicate that the SIKVAV-modified chitosan hydrogels markedly enhance the wound healing process in diabetic mice. This effect may be attributed to several mechanisms, including differentiation of fibroblasts, proliferation of keratinocytes, the promotion of angiogenesis, stimulation of collagen synthesis, upregulation of growth factor expression, and possible involvement of the TGF-β1/Smad3 signaling pathway. This research not only provides a new biomaterial for the treatment of diabetic wounds but also elucidates the related molecular mechanisms involved in wound healing of DFUs, offering valuable insights for future clinical applications. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>SIKVAV-modified chitosan hydrogels enhanced the contraction of skin wounds in diabetic mice. (<b>A</b>) Representative photographs of typical wounds among the control group, peptide group, chitosan group, and SIKVAV-modified chitosan group at 5, 7, and 14 days post-surgery; (<b>B</b>) statistical evaluation of the residual wound percentages in diabetic mice across the control group, peptide group, chitosan group, and SIKVAV-modified chitosan group (<span class="html-italic">n</span> = 3, * for <span class="html-italic">p</span> &lt; 0.05, and ** for <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 2
<p>SIKVAV-modified chitosan hydrogels expedited the expression of α-SMA in the skin wounds of diabetic mice. (<b>A</b>) Immunohistochemistry illustrating the expression of α-SMA in the skin wounds across the control group, peptide group, chitosan group, and peptide-modified chitosan group at 5, 7, and 14 days post-surgery (scale bar: 50 μm); (<b>B</b>) statistical evaluation of α-SMA in the skin wounds among the control group, peptide group, chitosan group, and peptide-modified chitosan group at 5, 7, and 14 days following surgery (<span class="html-italic">n</span> = 3, * for <span class="html-italic">p</span> &lt; 0.05, and ** for <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 3
<p>SIKVAV-modified chitosan hydrogels enhanced the proliferation of keratinocytes in the skin wounds of diabetic mice. (<b>A</b>) The levels of K6 expression in keratinocytes were evaluated in the control group, peptide group, chitosan group, and peptide-modified chitosan group using immunohistochemical techniques at 5, 7, and 14 days following surgery (scale bar: 50 μm). (<b>B</b>) The optical density of keratinocyte K6 was statistically assessed in the control group, the peptide group, the chitosan hydrogels group, and the peptide-modified chitosan hydrogels group at 5, 7, and 14 days post-surgery (<span class="html-italic">n</span> = 3, ** for <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 4
<p>SIKVAV-modified chitosan hydrogels enhanced angiogenesis in the skin wounds of diabetic mice. (<b>A</b>) Immunohistochemical evaluation of CD31 expression in vascular endothelial cells of the skin wounds at 5, 7, and 14 days post-surgery among the control group, the peptide group, the chitosan group, and the SIKVAV-modified chitosan group (scale bar: 50 μm); (<b>B</b>) statistical assessment of new blood capillaries among the control group, peptide group, chitosan group, and SIKVAV-modified chitosan group (<span class="html-italic">n</span> = 3, * for <span class="html-italic">p</span> &lt; 0.05, and ** for <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 5
<p>Masson trichrome staining demonstrated the presence of newly synthesized collagen at 5, 7, and 14 days post-treatment in the skin wounds of diabetic mice across the control group, the peptide group, the chitosan group, and the SIKVAV-modified chitosan group (scale bar: 50 μm).</p>
Full article ">Figure 6
<p>SIKVAV-modified chitosan hydrogels significantly increased the mRNA expression levels of growth factors in the skin wounds of diabetic mice at 5, 7, and 14 days post-treatment. This enhancement includes the mRNA levels of (<b>A</b>) VEGF, (<b>B</b>) bFGF, and (<b>C</b>) EGF (<span class="html-italic">n</span> = 3, ** for <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 7
<p>SIKVAV-modified chitosan hydrogels increased the expression levels of TGF-β1 (<b>A</b>) and Smad3 (<b>B</b>) mRNA in the skin wounds of diabetic mice at 5, 7, and 14 days post-initial treatment (<span class="html-italic">n</span> = 3, ** for <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 8
<p>After subcutaneous injection of lentivirus that interferes with the expression of TGF-β1, SIKVAV-modified chitosan hydrogels accelerated the expression levels of TGF-β1 (<b>A</b>) and Smad3 (<b>B</b>) mRNA in the skin wounds of diabetic mice at 5, 7, and 14 days following initial treatment (<span class="html-italic">n</span> = 3, ** for <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">
29 pages, 5701 KiB  
Article
Polysaccharide-Stabilized Semisolid Emulsion with Vegetable Oils for Skin Wound Healing: Impact of Composition on Physicochemical and Biological Properties
by Giovanna Araujo de Morais Trindade, Laiene Antunes Alves, Raul Edison Luna Lazo, Kamila Gabrieli Dallabrida, Jéssica Brandão Reolon, Juliana Sartori Bonini, Karine Campos Nunes, Francielle Pelegrin Garcia, Celso Vataru Nakamura, Fabiane Gomes de Moraes Rego, Roberto Pontarolo, Marcel Henrique Marcondes Sari and Luana Mota Ferreira
Pharmaceutics 2024, 16(11), 1426; https://doi.org/10.3390/pharmaceutics16111426 - 8 Nov 2024
Viewed by 563
Abstract
Background/Objectives: The demand for natural-based formulations in chronic wound care has increased, driven by the need for biocompatible, safe, and effective treatments. Natural polysaccharide-based emulsions enriched with vegetable oils present promising benefits for skin repair, offering structural support and protective barriers suitable for [...] Read more.
Background/Objectives: The demand for natural-based formulations in chronic wound care has increased, driven by the need for biocompatible, safe, and effective treatments. Natural polysaccharide-based emulsions enriched with vegetable oils present promising benefits for skin repair, offering structural support and protective barriers suitable for sensitive wound environments. This study aimed to develop and evaluate semisolid polysaccharide-based emulsions for wound healing, incorporating avocado (Persea gratissima) and blackcurrant (Ribes nigrum) oils (AO and BO, respectively). Both gellan gum (GG) and kappa-carrageenan (KC) were used as stabilizers due to their biocompatibility and gel-forming abilities. Methods: Four formulations were prepared (F1-GG-AO; F2-KC-AO; F3-GG-BO; F4-KC-BO) and evaluated for physicochemical properties, spreadability, rheology, antioxidant activity, occlusive and bioadhesion potential, biocompatibility, and wound healing efficacy using an in vitro scratch assay. Results: The pH values (4.74–5.06) were suitable for skin application, and FTIR confirmed excipient compatibility. The formulations showed reduced occlusive potential, pseudoplastic behavior with thixotropy, and adequate spreadability (7.13–8.47 mm2/g). Lower bioadhesion indicated ease of application and removal, enhancing user comfort. Formulations stabilized with KC exhibited superior antioxidant activity (DPPH scavenging) and fibroblast biocompatibility (CC50% 390–589 µg/mL) and were non-hemolytic. Both F2-KC-AO and F4-KC-BO significantly improved in vitro wound healing by promoting cell migration compared to other formulations. Conclusions: These findings underscore the potential of these emulsions for effective wound treatment, providing a foundation for developing skin care products that harness the therapeutic properties of polysaccharides and plant oils in a natural approach to wound care. Full article
(This article belongs to the Special Issue Dosage Form Design and Delivery Therapy for Skin Disorders)
Show Figures

Figure 1

Figure 1
<p>Flowchart of the formulation and characterization procedures. The preparation of the semisolid emulsion involves several sequential steps (<b>A</b>): weighing the individual components for the oil phase (OP) and aqueous phase (AP), heating each phase separately to 70 °C to ensure proper dissolution and mixing, combining the phases by gradually pouring the aqueous phase (AP) into the oil phase (OP) under constant stirring to form a uniform emulsion, and obtaining the final gel–cream formulation. The emulsion was subsequently characterized through various analyses (<b>B</b>): Fourier-transform infrared spectroscopy (FTIR) to assess molecular interactions and confirm compatibility among components, centrifugation to evaluate physical stability and detect any phase separation, spreadability and reology testing to determine ease of application and coverage on the skin, density measurement to assess formulation consistency, pH measurement with a pH meter to ensure suitability for skin application, bioadhesion and occlusion potential, antioxidant activity, cytotoxicity testing using cell cultures to evaluate biocompatibility and potential safety for skin use, and wound healing assay to determine efficacy.</p>
Full article ">Figure 2
<p>Macroscopic (<b>A</b>) and microscopic (<b>B</b>) images of polysaccharide-based semisolid emulsions containing vegetable oils. Overall, the formulations have a whitish color, homogeneous aspect, and shiny texture. The microscopic evaluation indicates that the system effectively dispersed the oil, keeping it stable within the semisolid structure. Abbreviations: GG—Gellan gum; KC—<span class="html-italic">Kappa</span>-carrageenan; BO—Blackcurrant Oil; AO—Avocado Oil.</p>
Full article ">Figure 3
<p>Infrared spectra of raw materials (<b>A</b>) and semisolid emulsions (<b>B</b>). The spectra exhibit characteristic peaks corresponding to the functional groups present in the substances. Additionally, these spectra support the compatibility among the components, as the absence of significant new peaks suggests no chemical interaction altering the molecular structure of the excipients.</p>
Full article ">Figure 4
<p>PCA model. In (<b>A</b>,<b>B</b>) are the eigenvalues graphs, which indicate that these three principal components encompass most of the chemical information in the raw materials. The red circles represent the principal components selected for the model. In (<b>C</b>,<b>D</b>) are the score plot graphs that reveal a distinct differentiation is observable between the formulations containing GG and KC, emphasizing these polysaccharides’ influence on the formulations’ ultimate chemical composition.</p>
Full article ">Figure 5
<p>Spreadability profile (<b>A</b>), spreadability factor (<b>B</b>), and viscosity (<b>C</b>) of semisolid emulsions. The developed emulsions demonstrated an increased spreading area with the application of more weight, suggesting they can expand more easily under pressure. Moreover, rheological measurements supported this behavior, as the complex viscosity (η*) of all formulations decreased with increasing angular frequency, which is a characteristic of pseudoplastic materials.</p>
Full article ">Figure 6
<p>Storage modulus (G′) and loss modulus (G″) as functions of angular frequency (ω). In (<b>A</b>,<b>B</b>) formulations containing AO stabilized with GG and KC, respectively. In (<b>C</b>,<b>D</b>) formulations prepared with BO stabilized with GG and KC, respectively. Data indicates that elastic and viscous behaviors become more pronounced at higher frequencies, suggesting a predominantly elastic rather than viscous behavior.</p>
Full article ">Figure 7
<p>Thixotropy evaluation of F1-GG-AO (<b>A</b>), F2-KC-AO (<b>B</b>), F3-GG-BO (<b>C</b>), and (<b>D</b>) F4-KC-BO. The data show that the material’s structure is temporarily disrupted under shear, but it recovers gradually when the shear is removed, which is characteristic of thixotropic materials.</p>
Full article ">Figure 8
<p>Antioxidant activity. The @ denotes the significant difference (<span class="html-italic">p</span> &lt; 0.05) between formulations and their respective blank forms (F1-GG-AO versusF5-GG-B, and F3-GG-BO versus F5-GG-B); # represents the significant difference (<span class="html-italic">p</span> &lt; 0.05) between polysaccharides (F1-GG-AO versus F2-KC-AO, and F5-GG-B versus F6-KC-B). NS means “not significant”. Both oils significantly enhanced the antioxidant potential of GG emulsions compared to the placebo semisolid, while emulsions stabilized with KC demonstrated higher antioxidant properties than those stabilized with GG.</p>
Full article ">Figure 9
<p>Occlusion potential. The @ denotes the significant difference (<span class="html-italic">p</span> &lt; 0.05) between formulations and their respective blank forms (F2-KC-AO versusF6-KC-B, and F4-KC-BO versus F6-KC-B); # represents the significant difference (<span class="html-italic">p</span> &lt; 0.05) between polysaccharides (F5-GG-B versus F6-KC-B). NS means “not significant”. Similar occlusion potential was observed among the formulations. Data also suggests that the oily components may negatively affect the KC formulations.</p>
Full article ">Figure 10
<p>Bioadhesion potential in intact and injured skin. The @ denotes the significant difference (<span class="html-italic">p</span> &lt; 0.05) between formulations and their respective blank forms; # represents the significant difference (<span class="html-italic">p</span> &lt; 0.05) between polysaccharides with the same oil (F1-GG-AO versusF2-KC-AO, or F3-GG-BO versus F4-KC-BO); * denotes the significant difference (<span class="html-italic">p</span> &lt; 0.05) between oils with the same polysaccharide (F1-GG-AO versus F3-GG-BO or F2-KC-AO versus F4-KC-BO); and <span>$</span> represents the significant difference (<span class="html-italic">p</span> &lt; 0.05) between intact and injured skin. NS means “not significant”. All formulations presented significantly higher bioadhesion in intact skin than in injured skin.</p>
Full article ">Figure 11
<p>Effect of F1-GG-AO (<b>A</b>), F2-KC-AO (<b>B</b>), F3-GG-BO (<b>C</b>), F4-KC-BO (<b>D</b>), F5-GG-B (<b>E</b>), and F6-KC-B (<b>F</b>) (1–1000 µg/mL) on the viability of L-929 cells by MTT assay. A negative control (non–treated cells) was conducted and considered 100% viability. Mean values were calculated from 3 independent results. The * denotes the significative difference from the negative control (<span class="html-italic">p</span> &lt; 0.05). NS means “not significant”. In all formulations examined, the viability of cells is observed to decline as the concentration increases.</p>
Full article ">Figure 12
<p>Hemolytic assay of KC semisolid emulsions. The results showed a hemolytic potential of less than 1% for all tested concentrations of the KC-based emulsions.</p>
Full article ">Figure 13
<p>Representative images showing the progression of healing over time (<b>A</b>) and percentage of open wound area at different times (0, 6, and 24 h) (<b>B</b>) for the F2-KC-AO, F4-KC-BO, and F6-KC-B, compared to the negative control. The * denotes the significant difference (<span class="html-italic">p</span> &lt; 0.05) with time zero in the same group, and # denotes the significant difference (<span class="html-italic">p</span> &lt; 0.05) with negative control at the same time. There is a consistent reduction in the area of open wounds over time, with formulations containing oils exhibiting a more pronounced degree of cell migration, which suggests an effective healing process.</p>
Full article ">
20 pages, 14484 KiB  
Article
Liver Fluke-Derived Molecules Accelerate Skin Repair Processes in a Mouse Model of Type 2 Diabetes Mellitus
by Anna Kovner, Yaroslav Kapushchak, Oxana Zaparina, Dmitry Ponomarev and Maria Pakharukova
Int. J. Mol. Sci. 2024, 25(22), 12002; https://doi.org/10.3390/ijms252212002 - 8 Nov 2024
Viewed by 387
Abstract
Chronic nonhealing wounds, such as diabetic ulcers, are among the most serious complications of diabetes mellitus. Consequently, the search for new therapeutic strategies remains highly relevant. Based on our previous data on acute wounds, bioactive molecules derived from the liver fluke Opisthorchis felineus [...] Read more.
Chronic nonhealing wounds, such as diabetic ulcers, are among the most serious complications of diabetes mellitus. Consequently, the search for new therapeutic strategies remains highly relevant. Based on our previous data on acute wounds, bioactive molecules derived from the liver fluke Opisthorchis felineus hold promise as a novel approach to wound healing. The aim of this study was to investigate the wound-healing properties of excretory–secretory products (ESP) and inactivated eggs of O. felineus in a model of type 2 diabetes mellitus. Two-month-old mice of the BKS.Cg + Leprdb/+Leprdb/OlaHsd (db/db) strain were inflicted with superficial wounds of 5 mm in diameter. Mouse groups included several controls (methylcellulose as the vehicle and human recombinant PDGF as the positive control) and specific-treatment groups (ESP and inactivated O. felineus eggs). Histopathological, immunohistochemical, and RT-PCR studies using markers for M1/M2 polarization, angiogenesis, and extracellular matrix remodeling were carried out. Additionally, an image analysis of Masson’s trichrome-stained skin sections was performed. The proliferation of HaCaT cells under ESP and egg treatment was also assessed. The present study reveals a significant increase in the percentage of wound healing in ESP- and egg-treated groups, which significantly exceeded the control values after 14 days. Wound treatment with either ESP or worm eggs resulted in (i) a reduction in inflammation with a canonical M1-to-M2 polarization shift, (ii) the modulation of the vascular response, and (iii) dermal extracellular matrix remodeling. All results are comparable to those of the positive control group treated with PDGF. This study also reveals that ESP, but not O. felineus eggs, stimulated keratinocyte proliferation in vitro. The results indicate the high wound-healing potential of liver fluke bioactive molecules and open prospects for further research on these new promising therapeutic approaches. Full article
(This article belongs to the Special Issue Cutaneous Biology, Molecular Dermatology and Dermatopathology)
Show Figures

Figure 1

Figure 1
<p>The pancreas and blood glucose in experimental db/db mice. (<b>A</b>) Representative pictures of pancreatic islets. Hematoxylin and eosin (H&amp;E) staining, magnification ×400. (<b>B</b>) Blood glucose levels (mmol/L). * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01.</p>
Full article ">Figure 2
<p>Effects of <span class="html-italic">O. felineus</span> ESP and inactivated eggs on diabetic wound healing. (<b>A</b>) Representative images of the wound on days 0, 4, 10, and 14 in several groups: without treatment, 1.5% methylcellulose (vehicle), positive control (PDGF), and specific treatment (ESP or Eggs). The wound area is delineated with a dotted line. (<b>B</b>) The percentage of wound area (* <span class="html-italic">p</span> &lt; 0.05 compared to the “no treatment” group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 compared to the “vehicle”; <sup><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.05 compared to the “PDGF”).</p>
Full article ">Figure 3
<p>Evaluation of the inflammation phase of wound healing. (<b>A</b>) Histopathological analysis of the area of an inflammatory infiltrate (mm<sup>2</sup>) is presented as a heat map. (<b>B</b>) mRNA levels of genes <span class="html-italic">Arg1</span> and <span class="html-italic">Nos2</span> were normalized to the mRNA level of <span class="html-italic">Hprt</span>. Data are presented as mean ± SEM, * <span class="html-italic">p</span> ≤ 0.05, *** <span class="html-italic">p</span> ≤ 0.001 compared to the vehicle group. (<b>C</b>) Representative histological images: wound area, H&amp;E staining, and 14 days after treatment, magnification ×40. The dotted line marks the wound area; IHC staining for arginase-1 and inducible NO synthase, 14 days after treatment, magnification ×200; stain-positive cells are indicated with arrows. (<b>D</b>) Histopathological analysis of the number of Arg1- and iNOS-positive cells is presented as a heat map. Data are presented as mean ± SEM, * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01, *** <span class="html-italic">p</span> ≤ 0.001 compared to the vehicle group; # <span class="html-italic">p</span> ≤ 0.05, ## <span class="html-italic">p</span> ≤ 0.01, ### <span class="html-italic">p</span> ≤ 0.001 compared to the PDGF group.</p>
Full article ">Figure 4
<p>Evaluation of the proliferation stage of wound healing in db/db mice. (<b>A</b>) Histopathological analysis of the number of CD31-positive (total) and CD34-positive (young and newly formed) vessels is presented as a heat map. (<b>B</b>) The mRNA level of the <span class="html-italic">Vegfa</span> gene was normalized to the mRNA level of <span class="html-italic">Hprt</span>. Data are presented as mean ± SEM, * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01, *** <span class="html-italic">p</span> ≤ 0.001 compared to the vehicle group; # <span class="html-italic">p</span> ≤ 0.05, ### <span class="html-italic">p</span> ≤ 0.001 compared to the PDGF group. (<b>C</b>) IHC staining for CD31 and CD34, 14 days after treatment, magnification ×200; stain-positive vessels are marked by arrows.</p>
Full article ">Figure 5
<p>Evaluation of the remodeling stage of wound healing in db/db mice. (<b>A</b>) Histopathological analysis of the connective-tissue proportion (%) in the wound area is presented as a heat map. (<b>B</b>) The mRNA level of <span class="html-italic">Tgfb1</span> was normalized to that of <span class="html-italic">Hprt</span>. Data are presented as mean ± SEM, * <span class="html-italic">p</span> ≤ 0.05 compared to the vehicle group. (<b>C</b>) Representative pictures of connective tissue in the wound area on the 14th day after treatment (Masson staining, collagen fibers are blue), IHC staining for type I collagen and α-smooth muscle actin; the stain-positive area is indicated by arrows); magnification ×100. (<b>D</b>) mRNA levels of genes <span class="html-italic">Col1a1</span>, <span class="html-italic">Mmp2</span>, and <span class="html-italic">Acta2</span> were normalized to the mRNA level of the <span class="html-italic">Hprt</span> gene. Data are presented as mean ± SEM, * <span class="html-italic">p</span> ≤ 0.05 compared to the vehicle group.</p>
Full article ">Figure 6
<p>Proliferation of HaCaT cells after treatment with ESP. (<b>A</b>) The number of HaCaT cells after incubation with <span class="html-italic">O. felineus</span> ESP in the medium with 1% of FBS. Seven replicates were implemented. Data are represented as mean ± SD. (<b>B</b>) HaCaT cells after 7 days of cultivation with <span class="html-italic">O. felineus</span> ESP, magnification ×100. (<b>C</b>) Immunocytochemical analysis of HaCaT cells. Cells stained for an <span class="html-italic">O. felineus</span> common antigen (green) and DAPI-labeled nuclei (blue), magnification ×400. NTC: no-treatment control cells; ESP: cells treated with ESP of <span class="html-italic">O. felineus</span>. * <span class="html-italic">p</span> &lt; 0.033 compared to the no-treatment control group; *** <span class="html-italic">p</span> &lt; 0.001 compared to the no-treatment control group.</p>
Full article ">Figure 7
<p>Schematic illustrating the potential cascade of wound-healing reactions of bioactive molecules from the liver fluke <span class="html-italic">Opisthorchis felineus</span> in a mouse model of type 2 diabetes mellitus (prepared using GIMP 2.10, <a href="https://www.gimp.org/" target="_blank">https://www.gimp.org/</a>). ↓: Indicates a decrease in the number of positive cells and/or a decrease in the expression of the specified gene. ↑: Indicates an increase in the number of positive cells and/or an increase in the expression of the specified gene.</p>
Full article ">Figure 8
<p>Schematic presentation of the experimental design. Each BKS.Cg + Leprdb/+Leprdb/OlaHsd (db/db) mouse (n = 48) was inflicted a superficial wound (5 mm in diameter). The entire duration of the experiment was 14 days, during which the animals were anesthetized twice and received sedation thrice. All groups received treatment every 3 days with simultaneous measurement of the wound area. Mice were withdrawn from the experiment on days 4, 10, and 14 of treatment with simultaneous collection of damaged-skin samples for histological examination and for gene expression analysis.</p>
Full article ">
14 pages, 6476 KiB  
Article
Conventional Versus Regenerative Methods for Wound Healing: A Comparative Experimental Study on a Sheep Model
by Rossella Elia, Michele Maruccia, Pietro Giovanni Di Summa, Rodrigo Trisciuzzi, Giuditta Lovero, Gerardo Cazzato, Luca Lacitignola, Francesco Staffieri and Alberto Maria Crovace
Medicina 2024, 60(11), 1836; https://doi.org/10.3390/medicina60111836 - 8 Nov 2024
Viewed by 439
Abstract
Background and Objectives: Wound healing is a complex process involving cellular, anatomical, and functional repair, often hindered in chronic wounds associated with diseases like diabetes and vascular disorders. This study investigated the efficacy of conventional and regenerative wound healing approaches in a [...] Read more.
Background and Objectives: Wound healing is a complex process involving cellular, anatomical, and functional repair, often hindered in chronic wounds associated with diseases like diabetes and vascular disorders. This study investigated the efficacy of conventional and regenerative wound healing approaches in a sheep surgical wound model. Materials and Methods: Six female Bergamasca sheep underwent five full-thickness skin lesions treated with various methods: sterile gauze (control), chlorhexidine, sodium hypochlorite, micronized dermis system application, and dermal matrix. Wound healing progression was monitored over 42 days through wound dimension measurements, exudate analysis, and histopathological evaluations. Results: The results indicated that all wounds healed completely by day 42, with significant reductions in wound size and exudate over time. Notably, Micronized dermis system application and dermal matrix treatments showed a faster evolution in exudate characteristics and improved collagen reorganization compared to other treatments. Histological analysis revealed earlier neovascularization and better reconstitution of hair follicles in these groups. Despite the lack of significant differences in healing time, both regenerative approaches enhanced wound healing phases, contributing to exudate control, angiogenesis promotion, and reduced scar formation. Conclusions: The findings suggest that while micronized dermis system application and dermal matrix do not accelerate acute wound healing compared to conventional methods, they offer potential benefits in managing exudate and improving tissue regeneration, warranting further investigation in chronic wound scenarios. Full article
Show Figures

Figure 1

Figure 1
<p>Control and experimental skin lesions produced on the backs of the sheep.</p>
Full article ">Figure 2
<p>Micronized dermis system application.</p>
Full article ">Figure 3
<p>Progressive wound contraction and healing over time. Wound diameter was evaluated at each time point with a caliper.</p>
Full article ">Figure 4
<p>Pictures refer to sheep n.1 and show the clinical appearance of the wounds belonging to different treatment methods (from A to E) at each time point.</p>
Full article ">Figure 5
<p>Histological photomicrograph of the healed tissue at T42 (sheep n.1). The image on the left belongs to the dermal matrix, while the image on the right belongs to the control area. The almost complete absence of dermal and subcutaneous infiltrate and granulation tissue could be observed (HE original magnification 20×).</p>
Full article ">Figure 6
<p>(<b>A</b>–<b>C</b>) Histological photomicrograph taken at T42 showing in (<b>A</b>) the reorganization of the collagen fibers in the DERMAL MATRIX group (HE original magnification 20× example in the black circle). (<b>B</b>) Another histological photomicrograph of the same DERMAL MATRIX group showing the reappearance of cutaneous adnexa (left, black circle) and the reorganization of the collagen fibers in the middle of the picture (HE original magnification 20×). (<b>C</b>) Histological photomicrograph showing the keratinization of the epidermis (HE original magnification 4×).</p>
Full article ">Figure 7
<p>Histological photomicrograph showing poor scarring with advanced fibrosis in the control area (HE original magnification 20×).</p>
Full article ">
15 pages, 592 KiB  
Review
Characterizing Fibroblast Heterogeneity in Diabetic Wounds Through Single-Cell RNA-Sequencing
by Helen H. Wang, Maria Korah, Serena L. Jing, Charlotte E. Berry, Michelle F. Griffin, Michael T. Longaker and Michael Januszyk
Biomedicines 2024, 12(11), 2538; https://doi.org/10.3390/biomedicines12112538 - 7 Nov 2024
Viewed by 548
Abstract
Diabetes mellitus is an increasingly prevalent chronic metabolic disorder characterized by physiologic hyperglycemia that, when left uncontrolled, can lead to significant complications in multiple organs. Diabetic wounds are common in the general population, yet the underlying mechanism of impaired healing in such wounds [...] Read more.
Diabetes mellitus is an increasingly prevalent chronic metabolic disorder characterized by physiologic hyperglycemia that, when left uncontrolled, can lead to significant complications in multiple organs. Diabetic wounds are common in the general population, yet the underlying mechanism of impaired healing in such wounds remains unclear. Single-cell RNA-sequencing (scRNAseq) has recently emerged as a tool to study the gene expression of heterogeneous cell populations in skin wounds. Herein, we review the history of scRNAseq and its application to the study of diabetic wound healing, focusing on how innovations in single-cell sequencing have transformed strategies for fibroblast analysis. We summarize recent research on the role of fibroblasts in diabetic wound healing and describe the functional and cellular heterogeneity of skin fibroblasts. Moreover, we highlight future opportunities in diabetic wound fibroblast research, with a focus on characterizing distinct fibroblast subpopulations and their lineages. Leveraging single-cell technologies to explore fibroblast heterogeneity and the complex biology of diabetic wounds may reveal new therapeutic targets for improving wound healing and ultimately alleviate the clinical burden of chronic wounds. Full article
(This article belongs to the Collection Feature Papers in Cell Biology and Pathology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Illustrated Workflow for scRNAseq of Diabetic Wounds. An example workflow for single-cell RNA-seq of diabetic skin wounds using a droplet-based method such as 10x Genomics Chromium, from specimen harvest to downstream analysis of sequencing data. Optional cell sorting and in silico cell type isolation steps are illustrated using forked arrows. Created in BioRender. Wang, H. (2024) <a href="http://BioRender.com/p83k591" target="_blank">BioRender.com/p83k591</a> (accessed on 11 October 2024).</p>
Full article ">
19 pages, 13454 KiB  
Article
Transcriptomics Revealed Differentially Expressed Transcription Factors and MicroRNAs in Human Diabetic Foot Ulcers
by Vikrant Rai
Proteomes 2024, 12(4), 32; https://doi.org/10.3390/proteomes12040032 - 5 Nov 2024
Viewed by 627
Abstract
Non-healing diabetic foot ulcers (DFUs) not only significantly increase morbidity and mortality but also cost a lot and drain healthcare resources. Persistent inflammation, decreased angiogenesis, and altered extracellular matrix remodeling contribute to delayed healing or non-healing. Recent studies suggest an increasing trend of [...] Read more.
Non-healing diabetic foot ulcers (DFUs) not only significantly increase morbidity and mortality but also cost a lot and drain healthcare resources. Persistent inflammation, decreased angiogenesis, and altered extracellular matrix remodeling contribute to delayed healing or non-healing. Recent studies suggest an increasing trend of DFUs in diabetes patients, and non-healing DFYs increase the incidence of amputation. Despite the current treatment with offloading, dressing, antibiotics use, and oxygen therapy, the risk of amputation persists. Thus, there is a need to understand the molecular and cellular factors regulating healing in DFUs. The ongoing research based on proteomics and transcriptomics has predicted multiple potential targets, but there is no definitive therapy to enhance healing in chronic DFUs. Increased or decreased expression of various proteins encoded by genes, whose expression transcriptionally and post-transcriptionally is regulated by transcription factors (TFs) and microRNAs (miRs), regulates DFU healing. For this study, RNA sequencing was conducted on 20 DFU samples of ulcer tissue and non-ulcerated nearby healthy tissues. The IPA analysis revealed various activated and inhibited transcription factors and microRNAs. Further network analysis revealed interactions between the TFs and miRs and the molecular targets of these TFs and miRs. The analysis revealed 30 differentially expressed transcription factors (21 activated and 9 inhibited), two translational regulators (RPSA and EIF4G2), and seven miRs, including mir-486, mir-324, mir-23, mir-186, mir-210, mir-199, and mir-338 in upstream regulators (p < 0.05), while causal network analysis (p < 0.05) revealed 28 differentially expressed TFs (19 activated and 9 inhibited), two translational regulators (RPSA and EIF4G2), and five miRs including mir-155, mir-486, mir-324, mir-210, and mir-1225. The protein–protein interaction analysis revealed the interaction of various novel proteins with the proteins involved in regulating DFU pathogenesis and healing. The results of this study highlight many activated and inhibited novel TFs and miRs not reported in the literature so far, as well as the targeted molecules. Since proteins are the functional units during biological processes, alteration of gene expression may result in different proteoforms and protein species, making the wound microenvironment a complex protein interaction (proteome complexity). Thus, investigating the effects of these TFs and miRs on protein expression using proteomics and combining these results with transcriptomics will help advance research on DFU healing and delineate potential therapeutic strategies. Full article
Show Figures

Figure 1

Figure 1
<p>TF-miRNA coregulatory interactions between activated and inhibited TFs and miRs in upstream regulators (IPA analysis): The literature-curated regulatory interaction information was collected from the RegNetwork repository using miRTarBase v9.0 (Inbuilt in Networkanalyst.ca). Blue squares—miRs, pink squares—newly appeared TFs, red/orange and yellow squares and circles—input TFs.</p>
Full article ">Figure 2
<p>TF-miRNA coregulatory interactions between activated and inhibited TFs and miRs in the causal network (IPA analysis): The literature-curated regulatory interaction information was collected from the RegNetwork repository using miRTarBase v9.0 (Inbuilt in Networkanalyst.ca). Blue squares—miRs, pink squares—newly appeared TFs, red/orange and yellow squares and circles—input TFs.</p>
Full article ">Figure 3
<p>Gene–transcription factors interaction network analysis using the Transcriptional Regulatory Relationships Unraveled by Sentence-based Text-mining (TRRUST) database of human transcriptional regulatory networks. Red circles—input genes (regulated by TFs in upstream regulator analysis), blue squares—TFs regulating these genes.</p>
Full article ">Figure 4
<p>Gene–transcription factors interaction network analysis using the Encyclopedia of DNA Elements (ENCODE) database. Red circles—input genes (regulated by TFs in upstream regulator analysis), blue squares—TFs regulating these genes.</p>
Full article ">Figure 5
<p>Gene–transcription factors interaction network analysis using Transcriptional Regulatory Relationships Unraveled by Sentence-based Text-mining (TRRUST) database of human transcriptional regulatory networks. Red circles—input genes (regulated by TFs in causal network analysis), blue squares—TFs regulating these genes.</p>
Full article ">Figure 6
<p>Gene–transcription factors interaction network analysis using the Encyclopedia of DNA Elements (ENCODE) database. Red circles—input genes (regulated by TFs in causal network analysis), blue squares—TFs regulating these genes.</p>
Full article ">Figure 7
<p>Network analysis revealing interaction between CXCL8 and S100 proteins. There was no direct interaction between CXCL8 and S100 proteins; however, there was an interaction of S100 proteins with CEBPB and IL-4, which in turn interacts with CXCL8 through NF-κB. This suggests that infiltration of immune cells and increased secretion of pro-inflammatory cytokines perpetuates inflammation, a major factor contributing to the non-healing of DFUs. S100 proteins are secreted by immune cells, which, on activation, secretes pro-inflammatory cytokines. Further, interaction of these proteins with inflammation, angiogenesis, differentiation, degranulation, phagocytosis, and T-cell activation suggests their role and the possibility of targeting them to promote DFU healing.</p>
Full article ">Figure 8
<p>STRING network for activated transcription factors CEBPB, CEBPA, ECSIT, EHF, E74 like ETS transcription factor 4 (ELF4), EP300, FOXL2, MAZ, NFE2L2, PPRC1, ZHX2, and ZIC5.</p>
Full article ">Figure 9
<p>STRING network for transcription factors BTG2, DLX1, ETV3, FOXA1, HIVEP1, KMT2D, NONO, WT1, and ZNF366.</p>
Full article ">
14 pages, 4579 KiB  
Article
Effective Treatment of Basal Cell Carcinoma with a Topical Enzymatic Mixture Enriched in Bromelain: Summary of Proof-Of Concept Clinical Studies on the First 22 Tumors
by Lior Rosenberg, Yaron Shoham, Brian Berman, Stephen K. Tyring, Michael D. Tharp and Adam J. Singer
J. Clin. Med. 2024, 13(21), 6624; https://doi.org/10.3390/jcm13216624 - 4 Nov 2024
Viewed by 876
Abstract
Background/Objectives: Basal cell carcinoma (BCC), the most prevalent form of human cancer, is traditionally treated by surgical and alternative destructive or topical chemical means, each with its advantages, challenges, and drawbacks. We describe our experience treating BCCs with a topical concentrate of proteolytic [...] Read more.
Background/Objectives: Basal cell carcinoma (BCC), the most prevalent form of human cancer, is traditionally treated by surgical and alternative destructive or topical chemical means, each with its advantages, challenges, and drawbacks. We describe our experience treating BCCs with a topical concentrate of proteolytic enzymes enriched in bromelain (CPEEB) sourced from pineapple stems. CPEEB has strong proteolytic, antitumor–proapoptotic, and inflammation modulation activities, and is approved for debridement of deep burns and starting phase 3 trials for chronic wounds. Methods: In the first proof-of-concept (POC) study, six BCCs on three individuals were treated with five to six daily CPEEB 10% topical applications under a zinc oxide-based occlusive dressing for 9–12 h each during a period of up to 10 days. These patients were followed for up to 4 years. In an additional two POC studies, 16 patients with one BCC each were treated every other day for a total of seven applications of topical CPEEB 5% under a variety of occlusive dressings. The wounds were followed for up to 2 months before undergoing diagnostic excisional biopsy. Results: In the first study, clinical assessment of the BCCs and two excisional biopsies after 6 months suggested that all lesions were eradicated with spontaneous healing within ~2 weeks without clinical or histological recurrence for over 4 years. In the two subsequent studies, 16 histologically diagnosed superficial and nodular BCCs were treated using four application techniques. Excisional histology after 2 months confirmed BCC eradication in seven of the patients. In nine patients, with compromised occlusive dressings, histological eradication was incomplete. Treatment was well tolerated by all patients with the expected local skin reactions, which completely healed within 2–3 weeks. Conclusions: These are POC preliminary studies aimed at indicating the potential efficacy and feasibility of topical CPEEB in eradicating BCC. In these studies, topical CPEEB 10% and 5% resulted in complete eradication of the BCC when appropriately applied. CPEEB was well tolerated in all patients, and all treated sites’ erosions healed without scars in <3 weeks. Further research is necessary to corroborate the results, refine the application technique, and complete the regulatory process. Full article
(This article belongs to the Section Oncology)
Show Figures

Figure 1

Figure 1
<p>Schematic description of the study design. In the first proof-of-concept (POC) study, the enrollment (1 day) and treatment phases were shorter (no diagnostic histology and only 5–6 daily treatments) compared to the second study. The post-treatment phases were longer in the first POC study (excisional biopsy after 6 months for two lesions and follow up for &gt;4 years) compared to the second study, where all underwent excisional biopsy after 2 months and follow-up until wound closure at 1 month. The third study, conducted in Israel, followed the same scheme as the second study in the US. The last two studies ended in wide-margin complete excisions without additional follow-up. The shaded area represents optional treatments that were only used on one of the lesions once (sixth application). TD: tumor destruction; FU: follow-up.</p>
Full article ">Figure 2
<p>Group 1 lesion. (<b>a</b>) Application of Aquaphor adhesive barrier and (<b>b</b>) coverage by Telfa.</p>
Full article ">Figure 3
<p>Group 1 lesion. (<b>a</b>) Nodular BCC pre-treatment, (<b>b</b>) post first treatment, (<b>c</b>) post treatment #2, (<b>d</b>) one day post treatment #3, (<b>e</b>) post treatment #4. (<b>f</b>) post treatment #5, (<b>g</b>) one day post treatment #6 and pre-treatment #7, (<b>h</b>) post treatment #7, (<b>i</b>) two months post last treatment, before excisional biopsy; see yellow arrow: clinical and histological clearing.</p>
Full article ">Figure 4
<p>Group 2 lesions. CPEEB dripping out, underneath Aquaphor irritating the exposed skin (<b>a</b>). SBCC pre treatment, (<b>b</b>) post 7 applications (arrow points to the lesion), (<b>c</b>) 3 weeks later, complete healing, complete clinical clearence but histologically incomplete clearance.</p>
Full article ">Figure 5
<p>Group 2 lesions CPEEB dripping out, underneath Aquaphor irritating the exposed skin (<b>a</b>). NBCC pre treatment, (<b>b</b>) post 6 applications (arrow points to the lesion) the CPEEB irritates the skin, belowand far from the lesion, (<b>c</b>) 4 weeks later, complete healing, complete clinical clearence but histologicly suspected residual BCC.</p>
Full article ">Figure 6
<p>Group 3 lesions. (<b>a</b>–<b>d</b>) The colostomy ring chambers are overfilled so that, in spite of some leakage (<b>b</b>,<b>c</b>) and dehydration (<b>e</b>–<b>g</b>), enough active CPEEB would remain to successfully clinically and histologically eradicate the BCC (<b>h</b>–<b>j</b>).</p>
Full article ">Figure 7
<p>Group 3 lesions. (<b>a</b>). NBCC post shave biopsy, (<b>b</b>). #1 treatment, colostomy ring dressing applied. (<b>c</b>). Post first treatment, erosion of the lesion and its surrounding with downword leakage, (<b>d</b>). #2 treatment, leakage under a colostomy ring with the expected irritation of the exposed skin below the treated lesion, (<b>e</b>). Decreased CPEEB dosedue to leakage with decreased effect. (<b>f</b>). One day post second treatment (<b>g</b>). Post #3 treatment, marked leakage with reduced effect. (<b>h</b>). One day after #5 treatment (<b>i</b>), One day post #6 treatment (<b>j</b>). One day post #7 treatment, (<b>k</b>). Good healing at 2 weeks and (<b>l</b>). Treated area after 2 months before excisional biopsy that was positive for suspected residual BCC in a scar. The arrows point to the treated tumor.</p>
Full article ">Figure 8
<p>Group 3 lesions. Evaporation of the CPEEB water through the occluding film. Dry CPEEB crusts are seen on the film’s inner surface in all applications (<b>a</b>–<b>g</b>). At the end of the treatment (<b>g</b>), with a minimal, insuficient effect on the target lesion after last application (<b>h</b>) and surrounding skin, and the final outcome after 6 weeks before excisional biopsy (<b>i</b>).</p>
Full article ">
Back to TopTop