Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Int J Nanomedicine ; 19: 2655-2673, 2024.
Article in English | MEDLINE | ID: mdl-38500680

ABSTRACT

Introduction: Immunotherapy has revolutionized cancer treatment by harnessing the immune system to enhance antitumor responses while minimizing off-target effects. Among the promising cancer-specific therapies, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has attracted significant attention. Methods: Here, we developed an ionizable lipid nanoparticle (LNP) platform to deliver TRAIL mRNA (LNP-TRAIL) directly to the tumor microenvironment (TME) to induce tumor cell death. Our LNP-TRAIL was formulated via microfluidic mixing and the induction of tumor cell death was assessed in vitro. Next, we investigated the ability of LNP-TRAIL to inhibit colon cancer progression in vivo in combination with a TME normalization approach using Losartan (Los) or angiotensin 1-7 (Ang(1-7)) to reduce vascular compression and deposition of extracellular matrix in mice. Results: Our results demonstrated that LNP-TRAIL induced tumor cell death in vitro and effectively inhibited colon cancer progression in vivo, particularly when combined with TME normalization induced by treatment Los or Ang(1-7). In addition, potent tumor cell death as well as enhanced apoptosis and necrosis was found in the tumor tissue of a group treated with LNP-TRAIL combined with TME normalization. Discussion: Together, our data demonstrate the potential of the LNP to deliver TRAIL mRNA to the TME and to induce tumor cell death, especially when combined with TME normalization. Therefore, these findings provide important insights for the development of novel therapeutic strategies for the immunotherapy of solid tumors.


Subject(s)
Colonic Neoplasms , Liposomes , Nanoparticles , Tumor Microenvironment , Animals , Mice , Ligands , Apoptosis , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Tumor Necrosis Factor-alpha , TNF-Related Apoptosis-Inducing Ligand/metabolism
2.
J Neuroimmunol ; 385: 578242, 2023 12 15.
Article in English | MEDLINE | ID: mdl-37951202

ABSTRACT

The pathophysiology of post-traumatic brain injury (TBI) behavioral and cognitive changes is not fully understood, especially in its mild presentation. We designed a weight drop TBI model in mice to investigate the role of neuroinflammation in behavioral and cognitive sequelae following mild TBI. C57BL/6 mice displayed depressive-like behavior at 72 h after mild TBI compared with controls, as indicated by a decrease in the latency to first immobility and climbing time in the forced swim test. Additionally, anxiety-like behavior and hippocampal-associated spatial learning and memory impairment were found in the elevated plus maze and in the Barnes maze, respectively. Levels of a set of inflammatory mediators and neurotrophic factors were analyzed at 6 h, 24 h, 72 h, and 30 days after injury in ipsilateral and contralateral hemispheres of the prefrontal cortex and hippocampus. Principal components analysis revealed two principal components (PC), which represented 59.1% of data variability. PC1 (cytokines and chemokines) expression varied between both hemispheres, while PC2 (neurotrophic factors) expression varied only across the investigated brain areas. Our model reproduces mild TBI-associated clinical signs and pathological features and might be a valuable tool to broaden the knowledge regarding mild TBI pathophysiology as well as to test potential therapeutic targets.


Subject(s)
Brain Concussion , Brain Injuries, Traumatic , Mice , Animals , Brain Concussion/complications , Mice, Inbred C57BL , Brain/pathology , Brain Injuries, Traumatic/complications , Nerve Growth Factors , Cognition , Maze Learning/physiology , Disease Models, Animal
3.
Toxicology ; 492: 153543, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37150288

ABSTRACT

This study aimed to evaluate the gold nanoparticles (AuNPs) animal sterilizing potential after intratesticular injections and long-term adverse reproductive and systemic effects. Adult male Wistar rats were divided into control and gold nanoparticle (AuNPs) groups. The rats received 200 µL of saline or AuNPs solution (16 µg/mL) on experimental days 1 and 7 (ED1 and ED7). After 150 days, the testicular blood flow was measured, and the rats were mated with females. After mating, male animals were euthanized for histological, cellular, and molecular evaluations. The female fertility indices and fetal development were also recorded. The results indicated increased blood flow in the testes of treated animals. Testes from treated rats had histological abnormalities, shorter seminiferous epithelia, and oxidative stress. Although the sperm concentration was lower in the AuNP-treated rats, there were no alterations in sperm morphology. Animals exposed to AuNPs had decreased male fertility indices, and their offspring had lighter and less efficient placentas. Additionally, the anogenital distance was longer in female fetuses. There were no changes in the histology of the kidney and liver, the lipid profile, and the serum levels of LH, testosterone, AST, ALT, ALP, albumin, and creatinine. The primary systemic effect was an increase in MDA levels in the liver and kidney, with only the liver experiencing an increase in CAT activity. In conclusion, AuNPs have a long-term impact on reproduction with very slight alterations in animal health. The development of reproductive biotechnologies that eliminate germ cells or treat local cancers can benefit from using AuNPs.


Subject(s)
Gold , Metal Nanoparticles , Pregnancy , Male , Female , Rats , Animals , Gold/toxicity , Rats, Wistar , Metal Nanoparticles/toxicity , Semen , Reproduction , Testis , Testosterone , Sperm Count , Sperm Motility , Spermatozoa
5.
Clin Sci (Lond) ; 137(7): 543-559, 2023 04 13.
Article in English | MEDLINE | ID: mdl-36972169

ABSTRACT

Poor disease outcomes and lethality are directly related to endothelial dysfunction in betacoronavirus infections. Here, we investigated the mechanisms underlying the vascular dysfunction caused by the betacoronaviruses MHV-3 and SARS-CoV-2. Wild-type C57BL/6 (WT) and knockout mice for inducible nitric oxide synthase (iNOS-/-) or TNF receptor 1 (TNFR1-/-) were infected with MHV-3, and K18-hACE2 transgenic mice expressing human ACE2 were infected with SARS-CoV-2. Isometric tension was used to evaluate vascular function. Protein expression was determined by immunofluorescence. Tail-cuff plethysmography and Doppler were used to assess blood pressure and flow, respectively. Nitric oxide (NO) was quantified with the DAF probe. ELISA was used to assess cytokine production. Survival curves were estimated using Kaplan-Meier. MHV-3 infection reduced aortic and vena cava contractility, arterial blood pressure, and blood flow, resulting in death. Resistance mesenteric arteries showed increased contractility. The contractility of the aorta was normalized by removing the endothelium, inhibiting iNOS, genetically deleting iNOS, or scavenging NO. In the aorta, iNOS and phospho-NF-kB p65 subunit expression was enhanced, along with basal NO production. TNF production was increased in plasma and vascular tissue. Genetic deletion of TNFR1 prevented vascular changes triggered by MHV-3, and death. Basal NO production and iNOS expression were also increased by SARS-CoV-2. In conclusion, betacoronavirus induces an endothelium-dependent decrease in contractility in macro-arteries and veins, leading to circulatory failure and death via TNF/iNOS/NO. These data highlight the key role of the vascular endothelium and TNF in the pathogenesis and lethality of coronaviruses.


Subject(s)
COVID-19 , Shock , Mice , Humans , Animals , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase/metabolism , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , SARS-CoV-2/metabolism , Mice, Inbred C57BL , Endothelium, Vascular/metabolism , Nitric Oxide/metabolism , Mice, Transgenic , Mesenteric Arteries/metabolism
6.
Reprod Sci ; 30(8): 2524-2536, 2023 08.
Article in English | MEDLINE | ID: mdl-36759496

ABSTRACT

Ovarian cryopreservation is an alternative for the preservation of fertility, and the subcutaneous transplantation site is considered one of the most promising. Studies evaluating the follicular growth and its relationship with gene expression and vascular perfusion are essential for improving this technique and its clinical application. Thus, the aim of this study was to evaluate the effect of subcutaneous autotransplantation and vitrification on follicular growth and atresia and their relationship with vascular perfusion and gene expression. Therefore, female mice were ovariectomized, and the ovaries were divided in two experimental groups (1) vitrified (treatment, n = 97) and (2) not vitrified (control, n = 97) and subsequently were transplanted. Then grafts, from both groups, were recovered after 1, 12, or 23 days (D1, D12, D23) and subjected to follicular quantification, morphometry, and qPCR. Non-transplanted ovaries (D0) were also used. The estrous cycle and vascular perfusion were monitored throughout the experiment. On D9, 100% of the animals had reestablished their estrous cycles (p > 0.05). Blood perfusion at the transplant site was similar for both treatments (p > 0.05), with greater perfusion at the site of vitrified transplants only on D1 (p < 0.05). A drastic reduction in the number of antral follicles and an increased number of atretic follicles were observed on D1 (p < 0.0001), associated with upregulation of Casp3, Fshr, and Igf1r; and downregulation of Bax, Acvr1, Egfr, and Lhcgr (p < 0.05). Our findings indicate that the first day after subcutaneous transplantation is a critical period for follicular survival, with intense follicular atresia independent of Bax upregulation.


Subject(s)
Follicular Atresia , Ovary , Female , Mice , Animals , bcl-2-Associated X Protein , Ovarian Follicle , Cryopreservation/methods , Vitrification , Gene Expression
7.
Acta Cir Bras ; 37(12): e371206, 2023.
Article in English | MEDLINE | ID: mdl-36651431

ABSTRACT

PURPOSE: To analyze the influence of occlusive dressing on the healing of excisional skin wounds in mice. METHODS: Pre-clinical, comparative, and translational study. Mice were divided into three experimental groups: wounds occluded with hydrocolloid (HD) dressings, transparent polyurethane film (TF) dressings, and without occlusion (WO), monitored at three, six and 14 days, with eight animals each. Closure rate, infiltration of neutrophils and macrophages, measurement of tumor necrosis factor-α (TNF-α) and vascular endothelial growth factor (VEGF) and, histologically, angiogenesis were evaluated. RESULTS: Wound closure was accelerated in the occlusive groups. There was a decrease in TNF-α levels in the HD group when compared to the WO and TF groups. Neutrophils accumulation decreased in the HD group. Increased dosages of macrophages were evidenced in the HD group, compared to the WO and TF groups. Levels of VEGF were increased in the TF and HD groups. CONCLUSIONS: It is suggested that the occlusion of wounds modulates the inflammatory response.


Subject(s)
Occlusive Dressings , Skin , Mice , Animals , Skin/pathology , Vascular Endothelial Growth Factor A , Tumor Necrosis Factor-alpha , Wound Healing/physiology , Models, Animal
8.
Acta Cir Bras ; 37(3): e370306, 2022.
Article in English | MEDLINE | ID: mdl-35674583

ABSTRACT

PURPOSE: To evaluate a biofilm model of Pseudomonas aeruginosa in excisional cutaneous wound in mice. METHODS: Preclinical, translational study conducted with 64 C57BL/6 mice randomly assigned to control and intervention groups. Evaluation was on days D0, D3, D5, D7 and D10 of wound making. The profile of biofilm formation and induction was evaluated using wound closure kinetics, quantitative culture, and evaluation of wounds using transmission electron microscopy (TEM). Clinical evaluation was performed by liver tissue culture, weight variation, and quantification of leukocytes in peripheral blood. Analyses were performed with GraphPad Prism software. RESULTS: Bacterial load for induction of infection with P. aeruginosa and survival of animals was 104 UFC·mL-1. In D5 (p < 0.0001) and D7 (p < 0.01), animals in the intervention group showed a delay in the healing process and had their wounds covered by necrotic tissue until D10. Statistical differences were observed in wound cultures and weight at D5 and D7 (p < 0.01). Liver cultures and leukocyte quantification showed no statistical differences. No bacteria in planktonic or biofilm form were identified by TEM. CONCLUSIONS: The findings raise questions about the understanding of the ease of formation and high occurrence of biofilm in chronic wounds.


Subject(s)
Pseudomonas Infections , Wound Infection , Animals , Mice , Biofilms , Mice, Inbred C57BL , Pseudomonas aeruginosa , Pseudomonas Infections/microbiology , Wound Infection/microbiology
9.
Arch Pharm (Weinheim) ; 355(9): e2200004, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35621705

ABSTRACT

For the first time, compounds developed from the 1,2,3-triazole scaffold were evaluated as novel drugs to treat triple-negative breast cancer (TNBC). Four organic salts were idealized as nonclassical bioisosteres of miltefosine, which is used in the topical treatment for skin metastasizing breast carcinoma. Among them, derivative dhmtAc displayed better solubility and higher cytotoxicity against the human breast adenocarcinoma cell line and mouse 4T1 cell lines, which are representatives of TNBC. In vitro assays revealed that dhmtAc interferes with cell integrity, confirmed by lactate dehydogenase leakage. Due to its human peripheral blood mononuclear cell (PBMC) toxicity, dhmtAc in vivo studies were carried out with the drug incorporated in a long-circulating and pH-sensitive liposome (SpHL-dhmtAc), and the acute toxicity in BALB/c mice was determined. Free dhmtAc displayed cardiac and pulmonary toxicity after the systemic administration of 5 mg/kg doses. On the other hand, SpHL-dhmtAc displayed no toxicity at 20 mg/kg. The in vivo antitumor effect of SpHL-dhmtAc was investigated using the 4T1 heterotopic murine model. Intravenous administration of SpHL-dhmtAc reduced the tumor volume and weight, without interfering with the body weight, compared with the control group and the dhmtAc free form. The incorporation of the triazole compound in the liposome allowed the demonstration of its anticancer potential. These findings evidenced 1,3,4-trisubstituted-1,2,3-triazole as a promising scaffold for the development of novel drugs with applicability for the treatment of patients with TNBC.


Subject(s)
Liposomes , Triple Negative Breast Neoplasms , Animals , Cell Line, Tumor , Humans , Leukocytes, Mononuclear , Mice , Mice, Inbred BALB C , Structure-Activity Relationship , Triazoles/pharmacology , Triple Negative Breast Neoplasms/drug therapy
10.
J Funct Biomater ; 13(2)2022 Apr 18.
Article in English | MEDLINE | ID: mdl-35466227

ABSTRACT

The quest for an ideal biomaterial perfectly matching the microenvironment of the surrounding tissues and cells is an endless challenge within biomedical research, in addition to integrating this with a facile and sustainable technology for its preparation. Engineering hydrogels through click chemistry would promote the sustainable invention of tailor-made hydrogels. Herein, we disclose a versatile and facile catalyst-free click chemistry for the generation of an innovative hydrogel by combining chondroitin sulfate (CS) and polyethylene glycol (PEG). Various multi-armed PEG-Norbornene (A-PEG-N) with different molecular sizes were investigated to generate crosslinked copolymers with tunable rheological and mechanical properties. The crosslinked and mechanically stable porous hydrogels could be generated by simply mixing the two clickable Tetrazine-CS (TCS) and A-PEG-N components, generating a self-standing hydrogel within minutes. The leading candidate (TCS-8A-PEG-N (40 kD)), based on the mechanical and biocompatibility results, was further employed as a scaffold to improve wound closure and blood flow in vivo. The hydrogel demonstrated not only enhanced blood perfusion and an increased number of blood vessels, but also desirable fibrous matrix orientation and normal collagen deposition. Taken together, these results demonstrate the potential of the hydrogel to improve wound repair and hold promise for in situ skin tissue engineering applications.

11.
Immunology ; 165(3): 355-368, 2022 03.
Article in English | MEDLINE | ID: mdl-34964126

ABSTRACT

Mucositis is a major clinical complication associated with cancer treatment and may limit the benefit of chemotherapy. Leukocytes and inflammatory mediators have been extensively associated with mucositis severity. However, the role of eosinophils in the pathophysiology of chemotherapy-induced mucositis remains to be elucidated. Here, using GATA-1-deficient mice, we investigated the role of eosinophils in intestinal mucositis. There was marked accumulation of eosinophils in mice given irinotecan and eosinophil ablation inhibited intestinal mucositis. Treatment with Evasin-4, a chemokine receptor antagonist, reduced the recruitment of eosinophils and decreased irinotecan-induced mucositis. Importantly, Evasin-4 did not interfere negatively with the antitumour effects of irinotecan. Evasin-4 was of benefit for mice given high doses of irinotecan once Evasin-4-treated mice presented delayed mortality. Altogether, our findings suggest that Evasin-4 may have significant mucosal-protective effects in the context of antineoplastic chemotherapy and may, therefore, be useful in combination with anticancer treatment in cancer patients.


Subject(s)
Antineoplastic Agents , Mucositis , Animals , Antineoplastic Agents/therapeutic use , Camptothecin/adverse effects , Eosinophils/pathology , Humans , Intestinal Mucosa/pathology , Irinotecan/adverse effects , Mice , Mucositis/chemically induced , Mucositis/drug therapy , Mucositis/pathology
12.
Estima (Online) ; 19(1): e1221, jan.-dez. 2021. tab, ilus
Article in English, Portuguese | BDENF - Nursing, LILACS | ID: biblio-1291466

ABSTRACT

Objetivo:validar método de fixação de curativos em feridas cutâneas excisionais de camundongos. Método: estudo pré-clínico. Amostra composta por animais da linhagem C57BL/6, que tiveram duas feridas excisionais confeccionadas na região dorsal. Foram avaliados diferentes métodos e produtos, amplamente aceitos na prática clínica, para fixação de curativos no modelo animal. Os desfechos avaliados foram tempo de permanência do curativo e ocorrência de eventos adversos. Resultados: atadura de crepom, fita microporosa e bandagem autoaderente apresentaram menor tempo de permanência quando comparadas ao filme de poliuretano. Esse, por sua vez, variou o tempo quando comparadas diferentes marcas (E, F, G e H) e número de voltas ao redor do corpo do animal. Com 1 volta, o tempo variou de < 24 a 36 horas. Com 2 voltas, as marcas E e G permaneceram 48 e 96 horas, respectivamente, e F e H tempo < 24 horas. Filme da marca G, cortado no tamanho 3 cm x 15 cm, dando 2 voltas no corpo do camundongo, manteve o curativo por 96 horas. A pele permaneceu íntegra, sem evento adverso. Conclusão: foi criado modelo de fixação de curativos para feridas em camundongos com produto disponível no Brasil e compatível com a estrutura copórea do animal.


Objective:validate method of fixation of dressings on excisional cutaneous wounds of mice. Method: preclinical study. Sample made up of animals of the C57BL/6 strain, which had two excision wounds made in the dorsal region. Different methods and products, widely accepted in clinical practice, for fixing dressings in the animal model were evaluated. The evaluated outcomes were the length of stay of the dressing and the occurrence of adverse events. Results: crepe bandage, microporous tape and self-adhesive bandage had a shorter residence time when compared to polyurethane film. This, in turn, varied the time when comparing different marks (E, F, G and H) and number of turns around the animal's body. With 1 lap, the time varied from <24 to 36 hours. With 2 laps, the marks E and G remained 48 and 96 hours, respectively, and F and H time <24 hours. G-brand film, cut to size 3 cm x 15 cm, giving the mouse body 2 turns, kept the dressing for 96 hours. The skin remained intact, with no adverse event. Conclusion: a dressing fixation model for wounds in mice was created with a product available in Brazil and compatible with the animal's body structure


Objetivo:validar método de fijación de apósitos en heridas cutáneas excisionales de ratones. Método: estudio preclínico. Muestra compuesta por animales del linaje C57BL/6 que tuvieron dos heridas excisionales confeccionadas en la región dorsal. Se evaluaron distintos métodos y productos, ampliamente aceptados en la práctica clínica, para fijación de apósitos en el modelo animal. Los resultados evaluados fueron tiempo de permanencia del apósito y ocurrencia de eventos adversos. Resultados: La venda de crepé, la cinta microporosa y el vendaje autoadherente presentaron menor tiempo de permanencia cuando comparados con la película de poliuretano. Esta, a su vez, varió en el tiempo cuando comparadas distintas marcas (E, F, G y H) y número de vueltas alrededor del cuerpo del animal. Con una vuelta completa, el tiempo varió de menos de 24 a 36 horas. Con dos vueltas, las marcas E y G permanecieron 48 y 96 horas, respectivamente, y F y H, tiempo igual e inferior a 24 horas. La piel permaneció íntegra, sin evento adverso. Conclusión: se creó un modelo de fijación de apósitos en ratones con un producto disponible en Brasil y compatible con la estructura del cuerpo del animal


Subject(s)
Bandages , Wound Healing , Basic Research
13.
Int Immunopharmacol ; 101(Pt B): 108224, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34655846

ABSTRACT

Jararhagin-C (Jar-C) is a disintegrin-like protein, isolated from the venom of B. jararaca, with affinity for α2ß1 integrin and the ability to incite processes such as angiogenesis and collagen deposition in vivo. Thus, we raised the hypothesis that this protein could be used as a therapeutic strategy for stimulating the healing of excisional wounds in mice. Four wounds were made on the back of Swiss mice, treated with daily intradermal injections of PBS (control group) or Jar-C (200 ng). Ten animals from each experimental group were euthanized and the tissue from the wounds and skin around them were collected for further biochemical, histological and molecular analysis. Wounds treated with Jar-C showed a faster closure rate, accompanied by a reduction in neutrophil infiltrate (MPO), pro-inflammatory cytokine levels (TNF, CXCL1 and CCL2) and an accumulation of macrophages in the analyzed tissues. It was also observed a greater expression of genes associated with the phenotype of alternatively activated macrophages (M2). Concomitantly, the administration of Jar-C holds an angiogenic potential, increasing the density of blood vessels and the synthesis of pro-angiogenic cytokines (VEGF and FGF). We also observed an increase in collagen deposition, accompanied by higher levels of the pro-fibrogenic cytokine TGF-ß1. Our data suggests Jar-C stimulates wound healing through stimulation of M2-like macrophage, angiogenesis and collagen deposition. Jar-C may be explored as a therapeutic strategy for wound healing, including the treatment of chronic wounds, where processes such as inflammation, angiogenesis and the deposition / remodeling of the matrix constituents are unregulated.


Subject(s)
Collagen , Crotalid Venoms , Disintegrins , Neovascularization, Physiologic , Wound Healing , Animals , Humans , Mice , Cell Survival/drug effects , Collagen/metabolism , Crotalid Venoms/chemistry , Cytokines/genetics , Cytokines/metabolism , Disintegrins/chemistry , Disintegrins/pharmacology , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells , Leukocytes/drug effects , Leukocytes/physiology , Macrophages , Neovascularization, Physiologic/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Wound Healing/drug effects , Bothrops jararaca Venom
14.
Life Sci ; 282: 119792, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34229006

ABSTRACT

AIMS: Exercise training increases circulating and tissue levels of angiotensin-(1-7) [Ang-(1-7)], which was shown to attenuate inflammation and fibrosis in different diseases. Here, we evaluated whether Ang-(1-7)/Mas receptor is involved in the beneficial effects of aerobic training in a chronic model of asthma. MATERIAL AND METHODS: BALB/c mice were subjected to a protocol of asthma induced by ovalbumin sensitization (OVA; 4 i.p. injections) and OVA challenge (3 times/week for 4 weeks). Simultaneously to the challenge period, part of the animals was continuously treated with Mas receptor antagonist (A779, 1 µg/h; for 28 days) and trained in a treadmill (TRE; 60% of the maximal capacity, 1 h/day, 5 days/week during 4 weeks). PGC1-α mRNA expression (qRT-PCR), plasma IgE and lung cytokines (ELISA), inflammatory cells infiltration (enzymatic activity assay) and airway remodeling (by histology) were evaluated. KEY FINDINGS: Blocking the Mas receptor with A779 increased IgE and IL-13 levels and prevented the reduction in extracellular matrix deposition in airways in OVA-TRE mice. Mas receptor blockade prevented the reduction of myeloperoxidase activity, as well as, prevented exercise-induced IL-10 increase. These data show that activation of Ang-(1-7)/Mas receptor pathway is involved in the anti-inflammatory and anti-fibrotic effects of aerobic training in an experimental model of chronic asthma. SIGNIFICANCE: Our results support exercise training as a non-pharmacological tool to defeat lung remodeling induced by chronic pulmonary inflammation. Further, our result also supports development of new therapy based on Ang-(1-7) or Mas agonists as important tool for asthma treatment in those patients that cannot perform aerobic training.


Subject(s)
Angiotensin I/metabolism , Asthma/therapy , Peptide Fragments/metabolism , Pneumonia/therapy , Angiotensin I/blood , Animals , Asthma/blood , Asthma/metabolism , Disease Models, Animal , Exercise Therapy , Male , Mice, Inbred BALB C , Peptide Fragments/blood , Pneumonia/blood , Pneumonia/metabolism
15.
Probiotics Antimicrob Proteins ; 13(3): 709-719, 2021 06.
Article in English | MEDLINE | ID: mdl-33433898

ABSTRACT

Skin wounds are an important clinical problem which affects millions of people worldwide. The search for new therapeutic approaches to improve wound healing is needed. The present study aimed to evaluate the effects of the oral treatment with the skin-related probiotics Lactobacillus johnsonii LA1 (LJ), L. paracasei ST11 (LP), and L. rhamnosus LPR (LR) in a model of excisional skin wounds in Swiss mice. The animals received daily oral gavage of PBS or 1 × 107 colony-forming units of LJ, LP, or LR, singly, beginning just after the creation of wounds until euthanasia. Blood flow was evaluated by laser Doppler perfusion imaging. Myeloperoxidase and N-acetyl-ß-D-glucosaminidase activities were used to assess the accumulation of neutrophils and macrophages, respectively. The wound tissue was also collected for histological analyses (H&E, Toluidine blue, and Picrosirius red staining). The macroscopic wound closure rate was faster only in mice treated with LR, but not with LJ and LP, when compared to mice treated with PBS. Histological evaluations showed that treatment with LR stimulated wound epithelization when compared to PBS. Further analyses showed that wounds from LR-treated mice presented a significant decrease in macrophage (p < 0.001) and mast cell (p < 0.001) infiltration, along with improved angiogenesis (p < 0.001) and blood flow (p < 0.01). Of note, collagen deposition and scarring were reduced in LR-treated mice when compared to PBS-treated mice. In conclusion, our results show that the oral treatment with Lactobacillus rhamnosus accelerates skin wound closure and reduces scar, besides to reducing inflammation and fibrogenesis and improving angiogenesis in the wounded skin.


Subject(s)
Cicatrix , Lacticaseibacillus rhamnosus , Probiotics/therapeutic use , Skin/injuries , Wound Healing , Animals , Cicatrix/prevention & control , Mice
16.
Am J Physiol Heart Circ Physiol ; 320(3): H1066-H1079, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33356962

ABSTRACT

Butyrate is a short-chain fatty acid (SCFA) derived from microbiota and is involved in a range of cell processes in a concentration-dependent manner. Low concentrations of sodium butyrate (NaBu) were shown to be proangiogenic. However, the mechanisms associated with these effects are not yet fully known. Here, we investigated the contribution of the SCFA receptor GPR43 in the proangiogenic effects of local treatment with NaBu and its effects on matrix remodeling using the sponge-induced fibrovascular tissue model in mice lacking the Gpr43 gene (Gpr43-KO) and the wild-type (WT) mice. We demonstrated that NaBu (0.2 mM intraimplant) treatment enhanced the neovascularization process, blood flow, and VEGF levels in a GPR43-dependent manner in the implants. Moreover, NaBu was able to modulate matrix remodeling aspects of the granulation tissue such as proteoglycan production, collagen deposition, and α-smooth muscle actin (α-SMA) expression in vivo, besides increasing transforming growth factor (TGF)-ß1 levels in the fibrovascular tissue, in a GPR43-dependent manner. Interestingly, NaBu directly stimulated L929 murine fibroblast migration and TGF-ß1 and collagen production in vitro. GPR43 was found to be expressed in human dermal fibroblasts, myofibroblasts, and endothelial cells. Overall, our findings evidence that the metabolite-sensing receptor GPR43 contributes to the effects of low dose of NaBu in inducing angiogenesis and matrix remodeling during granulation tissue formation. These data provide important insights for the proposition of new therapeutic approaches based on NaBu, beyond the highly explored intestinal, anti-inflammatory, and anticancer purposes, as a local treatment to improve tissue repair, particularly, by modulating granulation tissue components.NEW & NOTEWORTHY Our data show the contribution of the metabolite-sensing receptor GPR43 in the effects of low dose of sodium butyrate (NaBu) on stimulating angiogenesis and extracellular matrix remodeling in a model of granulation tissue formation in mice. We also show that human dermal fibroblasts, myofibroblasts, and endothelial cells express the receptor GPR43. These data provide important insights for the use of NaBu in local therapeutic approaches applicable to tissue repair in sites other than the intestine.


Subject(s)
Angiogenesis Inducing Agents/administration & dosage , Butyric Acid/administration & dosage , Extracellular Matrix/drug effects , Fibroblasts/drug effects , Granulation Tissue/drug effects , Neovascularization, Physiologic/drug effects , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Collagen/metabolism , Disease Models, Animal , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Granulation Tissue/metabolism , Granulation Tissue/pathology , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Surgical Sponges , Transforming Growth Factor beta1/metabolism
17.
Int Immunopharmacol, v. 101, 108224, dez. 2021
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: bud-3973

ABSTRACT

Jararhagin-C (Jar-C) is a disintegrin-like protein, isolated from the venom of B. jararaca, with affinity for α2β1 integrin and the ability to incite processes such as angiogenesis and collagen deposition in vivo. Thus, we raised the hypothesis that this protein could be used as a therapeutic strategy for stimulating the healing of excisional wounds in mice. Four wounds were made on the back of Swiss mice, treated with daily intradermal injections of PBS (control group) or Jar-C (200 ng). Ten animals from each experimental group were euthanized and the tissue from the wounds and skin around them were collected for further biochemical, histological and molecular analysis. Wounds treated with Jar-C showed a faster closure rate, accompanied by a reduction in neutrophil infiltrate (MPO), pro-inflammatory cytokine levels (TNF, CXCL1 and CCL2) and an accumulation of macrophages in the analyzed tissues. It was also observed a greater expression of genes associated with the phenotype of alternatively activated macrophages (M2). Concomitantly, the administration of Jar-C holds an angiogenic potential, increasing the density of blood vessels and the synthesis of pro-angiogenic cytokines (VEGF and FGF). We also observed an increase in collagen deposition, accompanied by higher levels of the pro-fibrogenic cytokine TGF-β1. Our data suggests Jar-C stimulates wound healing through stimulation of M2-like macrophage, angiogenesis and collagen deposition. Jar-C may be explored as a therapeutic strategy for wound healing, including the treatment of chronic wounds, where processes such as inflammation, angiogenesis and the deposition / remodeling of the matrix constituents are unregulated.

18.
Sci Rep ; 10(1): 19257, 2020 11 06.
Article in English | MEDLINE | ID: mdl-33159113

ABSTRACT

Photobiomodulation is being widely applied for improving dermal or mucosal wound healing. However, the underlying cellular and molecular processes that directly contribute to its effects remain poorly understood. Pericytes are relevant cells involved in the wound microenvironment and could be one of the main targets of photobiomodulation due to their plasticity and perivascular localization. Herein, we investigate tissue repair under the photobiomodulation stimulus using a pericyte labeled (or reporter) transgenic mice. Using a model of two contralateral back wounds, one the control and the other photoactivated daily (660 nm, 20 mW, 0.71 W/cm2, 5 J/cm2, 7 s, 0.14 J), we showed an overall influx of immune and undifferentiated cells and higher mobilization of a potent pericyte subpopulation (Type-2 pericytes) in the photoactivated wounds in comparison to the controls. Doppler analysis showed a significant increase in the blood flow in the photoactivated wounds, while marked vascular supply was observed histologically. Histochemical analysis has indicated more advanced stages of tissue repair after photoactivation. These data suggest that photobiomodulation significantly accelerates tissue repair through its vascular effects with direct recruitment of pericytes to the injury site.


Subject(s)
Low-Level Light Therapy , Pericytes/metabolism , Skin/injuries , Skin/metabolism , Wound Healing , Animals , Mice , Mice, Transgenic , Pericytes/pathology , Skin/pathology
19.
Immunobiology ; 225(3): 151957, 2020 05.
Article in English | MEDLINE | ID: mdl-32517880

ABSTRACT

Asthma is characterized by inflammation, pulmonary remodeling and bronchial hyperresponsiveness. We have previously shown that treatment with angiotensin-(1-7) [Ang-(1-7)] promotes resolution of eosinophilic inflammation and prevents chronic allergic lung inflammation. Here, we evaluated the effect of treatment with the inclusion compound of Ang-(1-7) in hydroxypropyl ß-cyclodextrin (HPßCD) given by inhalation on pulmonary remodeling in an ovalbumin (OVA)-induced chronic allergic lung inflammation. Mice were sensitized to ovalbumin (OVA; 4 injections over 42 days, 14 days apart) and were challenged 3 times per week, for 4 weeks (days 21-46). After the 2nd week of challenge, mice were treated with Ang-(1-7) by inhalation (4.5 µg of Ang-(1-7) included in 6.9 µg of HPßCD for 14 days, i.e. days 35-48). Mice were killed 72 h after the last challenge and blood, bronchoalveolar lavage fluid (BALF) and lungs were collected. Histology and morphometric analysis were performed in the lung. Metalloproteinase (MMP)-9 and MMP-12 expression and activity, IL-5, CCL11 in the lung and plasma IgE were measured. After 2 weeks of OVA challenge there was an increase in plasma IgE and in inflammatory cells infiltration in the lung of asthmatic mice. Treatment with inhaled administration of Ang-(1-7)/HPßCD for 14 days reduced eosinophils, IL5, CCL11 in the lung and plasma IgE. Treatment of asthmatic mice with Ang-(1-7)/HPßCD by inhalation reversed pulmonary remodeling by reducing collagen deposition and MMP-9 and MMP-12 expression and activity. These results show for the first time that treatment by inhalation with Ang-(1-7) can reverse an installed asthma, inhibiting pulmonary inflammation and remodeling.


Subject(s)
Airway Remodeling/drug effects , Angiotensin I/administration & dosage , Asthma/drug therapy , Asthma/metabolism , Peptide Fragments/administration & dosage , Vasodilator Agents/administration & dosage , Administration, Inhalation , Airway Remodeling/immunology , Animals , Asthma/diagnosis , Asthma/etiology , Biomarkers , Cytokines , Disease Models, Animal , Immunoglobulin E/blood , Immunoglobulin E/immunology , Lung/drug effects , Lung/immunology , Lung/pathology , Matrix Metalloproteinases/metabolism , Mice , Ovalbumin/adverse effects
20.
Atherosclerosis ; 304: 30-38, 2020 07.
Article in English | MEDLINE | ID: mdl-32574829

ABSTRACT

BACKGROUND AND AIMS: Peripheral arterial disease (PAD) is an important cause of morbidity and mortality with little effective medical treatment currently available. Nitric oxide (NO) is crucially involved in organ perfusion, tissue protection and angiogenesis. METHODS: We hypothesized that a novel NO-donor, MPC-1011, might elicit vasodilation, angiogenesis and arteriogenesis and in turn improve limb perfusion, in a hindlimb ischemia model. Hindlimb ischemia was induced by femoral artery ligation in Sprague-Dawley rats, which were randomized to receive either placebo, MPC-1011, cilostazol or both, up to 28 days. Limb blood flow was assessed by laser Doppler imaging. RESULTS: After femoral artery occlusion, limb perfusion in rats receiving MPC-1011 alone or in combination with cilostazol was increased throughout the treatment regimen. Capillary density and the number of arterioles was increased only with MPC-1011. MPC-1011 improved vascular remodeling by increasing luminal diameter in the ischemic limb. Moreover, MPC-1011 stimulated the release of proangiogenic cytokines, including VEGF, SDF1α and increased tissue cGMP levels, reduced platelet activation and aggregation, potentiated proliferation and migration of endothelial cells which was blunted in the presence of soluble guanylyl cyclase inhibitor LY83583. In MPC-1011-treated rats, Lin-/CD31+/CXCR4+ cells were increased by 92.0% and Lin-/VEGFR2+/CXCR4+ cells by 76.8% as compared to placebo. CONCLUSIONS: Here we show that the NO donor, MPC-1011, is a specific promoter of angiogenesis and arteriogenesis in a hindlimb ischemia model in an NO-cGMP-VEGF- dependent manner. This sets the basis to evaluate and confirm the efficacy of such therapy in a clinical setting in patients with PAD and impaired limb perfusion.


Subject(s)
Chemokine CXCL12 , Ischemia/drug therapy , Neovascularization, Physiologic/drug effects , Nitric Oxide Donors/pharmacology , Vascular Endothelial Growth Factor A , Animals , Disease Models, Animal , Endothelial Cells , Hindlimb , Muscle, Skeletal , Rats , Rats, Sprague-Dawley , Regional Blood Flow , Vascular Endothelial Growth Factor A/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...