1
|
Pinheiro JLS, Sousa WM, Rodrigues LHM, Bezerra FF, Cunha CLOA, Santos VMR, Oliveira SRBD, Bingana RD, Barbosa ALR, Souza MHLP, Freitas ALP, Damasceno ROS. Iota-Carrageenan from Marine Alga Solieria filiformis Prevents Naproxen-Induced Gastrointestinal Injury via Its Antioxidant and Anti-Inflammatory Activities. Biomedicines 2024; 12:2574. [PMID: 39595140 PMCID: PMC11592226 DOI: 10.3390/biomedicines12112574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 09/12/2024] [Accepted: 09/15/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Non-steroidal anti-inflammatory drugs (NSAIDs) are widely used in therapy due to their anti-inflammatory and analgesic properties. However, their clinical use is often associated with gastrointestinal complications. Thus, this study aimed to investigate the protective effect of a sulfated iota-carrageenan isolated from the marine alga Solieria filiformis (IC-Sf) against naproxen-induced gastrointestinal injury. Methods: Parameters of gastrointestinal injury, secretory and motor functions, and toxicity were evaluated. Results: The results demonstrated that IC-Sf significantly reduced naproxen-induced gastrointestinal macroscopic injury, with a maximum effect observed at 30 mg/kg. IC-Sf also preserved gastrointestinal antioxidant defense and prevented lipid peroxidation, with a reduction in the non-protein sulfhydryl group (NP-SH) and malondialdehyde (MDA) concentrations induced by naproxen. Additionally, IC-Sf mitigated naproxen-induced gastrointestinal inflammation, as evidenced by reduced myeloperoxidase (MPO) activity, tumor necrosis factor-alpha (TNF-α), and interleukin-1 beta (IL-1β). IC-Sf did not alter gastric secretion or gastrointestinal motility. In addition, the animals treated with IC-Sf did not present toxic effects. Conclusions: In conclusion, IC-Sf protected the gastrointestinal tract against the harmful effects of naproxen by inhibiting the inflammatory response and lipid peroxidation, suggesting its potential as a new therapeutic agent or food additive.
Collapse
Affiliation(s)
- João L. S. Pinheiro
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife 50670-420, PE, Brazil; (J.L.S.P.); (L.H.M.R.); (C.L.O.A.C.); (V.M.R.S.)
| | - Willer M. Sousa
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza 60020-181, CE, Brazil; (W.M.S.); (F.F.B.); (A.L.P.F.)
| | - Lucas H. M. Rodrigues
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife 50670-420, PE, Brazil; (J.L.S.P.); (L.H.M.R.); (C.L.O.A.C.); (V.M.R.S.)
| | - Francisco F. Bezerra
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza 60020-181, CE, Brazil; (W.M.S.); (F.F.B.); (A.L.P.F.)
| | - Cecília L. O. A. Cunha
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife 50670-420, PE, Brazil; (J.L.S.P.); (L.H.M.R.); (C.L.O.A.C.); (V.M.R.S.)
| | - Victória M. R. Santos
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife 50670-420, PE, Brazil; (J.L.S.P.); (L.H.M.R.); (C.L.O.A.C.); (V.M.R.S.)
| | - Samara R. B. D. Oliveira
- Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza 60430-275, CE, Brazil; (S.R.B.D.O.); (R.D.B.); (M.H.L.P.S.)
| | - Rudy D. Bingana
- Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza 60430-275, CE, Brazil; (S.R.B.D.O.); (R.D.B.); (M.H.L.P.S.)
| | - André Luiz. R. Barbosa
- Department of Physiotherapy, Parnaíba Delta Federal University, Parnaíba 64202-020, PI, Brazil;
| | - Marcellus H. L. P. Souza
- Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza 60430-275, CE, Brazil; (S.R.B.D.O.); (R.D.B.); (M.H.L.P.S.)
| | - Ana Lúcia P. Freitas
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza 60020-181, CE, Brazil; (W.M.S.); (F.F.B.); (A.L.P.F.)
| | - Renan O. S. Damasceno
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Recife 50670-420, PE, Brazil; (J.L.S.P.); (L.H.M.R.); (C.L.O.A.C.); (V.M.R.S.)
| |
Collapse
|
2
|
Keller CL, Jones NT, Abadie RB, Barham W, Behara R, Patil S, Paladini A, Ahmadzadeh S, Shekoohi S, Varrassi G, Kaye AD. Non-steroidal Anti-inflammatory Drug (NSAID)-, Potassium Supplement-, Bisphosphonate-, and Doxycycline-Mediated Peptic Ulcer Effects: A Narrative Review. Cureus 2024; 16:e51894. [PMID: 38333496 PMCID: PMC10849936 DOI: 10.7759/cureus.51894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/08/2024] [Indexed: 02/10/2024] Open
Abstract
Peptic ulcers are a common condition that arises from an imbalance between acid production and gastroduodenal protective factors. Various drugs, including non-steroidal anti-inflammatory drugs (NSAIDs), potassium supplements, bisphosphonates, and doxycycline, can increase the development of peptic ulcers. NSAIDs are one of the most common medications prescribed for pain relief, and they also inhibit the formation of cyclooxygenase-1 (COX-1). COX-1 helps in the production of mucus that lines the stomach, so by inhibiting COX-1, NSAIDs reduce the mucus produced by the stomach and increase the likelihood of gastric ulcer formation. Additionally, NSAIDs are acidic, and increasing the amount of any acid in the stomach can result in promoting ulcer development. Potassium supplements are used to reduce the effects of hypertension, decrease the development of kidney stones, and treat hypokalemia. The various types of transporters and channels used to move potassium across cell membranes increase hydrogen being pumped, increasing gastric acid production and ulcer formation. Bisphosphonates are used to treat a variety of skeletal disorders that require inhibition of osteoclast activity. Nitric oxide (NO) has been shown to have a therapeutic effect on gastric ulcers, and some bisphosphonates have been shown to decrease the production of nitric oxide, resulting in increased damage to the gastric mucosa. Finally, doxycycline is a broad-spectrum tetracycline antibiotic that is typically used to treat anthrax poisoning, skin lesions, and sexually transmitted diseases. A harmful adverse effect of doxycycline is the formation of peptic and gastric ulcers related to the drug being highly acidic once it has dissolved.
Collapse
Affiliation(s)
- Camryn L Keller
- School of Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Nicholas T Jones
- School of Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Raegan B Abadie
- School of Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - William Barham
- School of Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Raju Behara
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Shilpadevi Patil
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Antonella Paladini
- Department of Life, Health and Environmental Sciences (MESVA), University of L'Aquila, L'Aquila, ITA
| | - Shahab Ahmadzadeh
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Sahar Shekoohi
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | | | - Alan D Kaye
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| |
Collapse
|
3
|
Pessoa RT, Alcântara IS, da Silva LYS, da Costa RHS, Silva TM, de Morais Oliveira-Tintino CD, Ramos AGB, de Oliveira MRC, Martins AOBPB, de Lacerda BCGV, de Andrade EM, Ribeiro-Filho J, Gonçalves Lima CM, Coutinho HDM, Menezes IRAD. Ximenia americana L.: Chemical Characterization and Gastroprotective Effect. ANALYTICA 2023; 4:141-158. [DOI: 10.3390/analytica4020012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025] Open
Abstract
Ximenia americana L., popularly known in Brazil as “ameixa do-mato, ameixa-brava, and ameixa-do-sertão,” is widely used in folk medicine to treat several intestinal disorders. The present study assessed the potential mechanisms of action underlying the gastroprotective activity of the hydroethanolic extract of Ximenia americana L. (EHXA) stem bark. The acute toxicity of EHXA was estimated, and later, the gastroprotective effect in mice was assessed through acute models of gastric lesions induced by acidified or absolute ethanol and indomethacin, where the following mechanisms were pharmacologically analyzed: the involvement of prostaglandins (PG), histamine (H2) receptors, ATP-dependent potassium channels, sulfhydryl groups (SH), α2 adrenergic receptors, nitric oxide (NO), myeloperoxidase (MPO), gastric mucus production, and acetylcholine-mediated intestinal motility. Regarding toxicity, EHXA did not cause deaths or signs of toxicity (LD50 greater than or equal to 2000 mg/kg/p.o.). When the gastroprotective effect was assessed, EHXA (50, 100, and 200 mg/kg/p.o.) reduced the rate of lesions induced by acidified ethanol by 65.63; 53.66, and 58.02% in absolute ethanol at 88.91, 78.82, and 74.68%, respectively, when compared to the negative control group. In the indomethacin-induced gastric injury model, the reductions were 84.69, 55.99, 55.99, and 42.50%, respectively. The study revealed that EHXA might stimulate mucus production and reduce intestinal motility through SH groups, NO production, and activation of α2 adrenergic receptors. The results indicated that EHXA had significant gastroprotective activity in the evaluated models. However, further investigation is required to elucidate the cellular and molecular events underlying the action of EHXA components and to correlate them with the modulation of the signaling pathways, as demonstrated by the current pharmacological approach. Therefore, the results demonstrated in the present study, as well as previously reported findings, support the recommendation of using this species in traditional communities in Brazil.
Collapse
Affiliation(s)
- Renata Torres Pessoa
- Laboratory of Pharmacology and Molecular Chemistry, Department of Biological Chemistry, Regional University of Cariri, Rua Coronel Antônio Luis 1161, Crato 63105-000, CE, Brazil
| | - Isabel Sousa Alcântara
- Laboratory of Pharmacology and Molecular Chemistry, Department of Biological Chemistry, Regional University of Cariri, Rua Coronel Antônio Luis 1161, Crato 63105-000, CE, Brazil
| | - Lucas Yure Santos da Silva
- Laboratory of Pharmacology and Molecular Chemistry, Department of Biological Chemistry, Regional University of Cariri, Rua Coronel Antônio Luis 1161, Crato 63105-000, CE, Brazil
| | - Roger Henrique Souza da Costa
- Laboratory of Pharmacology and Molecular Chemistry, Department of Biological Chemistry, Regional University of Cariri, Rua Coronel Antônio Luis 1161, Crato 63105-000, CE, Brazil
| | - Tarcísio Mendes Silva
- Laboratory of Pharmacology and Molecular Chemistry, Department of Biological Chemistry, Regional University of Cariri, Rua Coronel Antônio Luis 1161, Crato 63105-000, CE, Brazil
| | - Cícera Datiane de Morais Oliveira-Tintino
- Laboratory of Microbiology and Molecular Biology, Department of Biological Chemistry, Regional University of Cariri, Rua Coronel Antônio Luis 1161, Crato 63105-000, CE, Brazil
| | - Andreza Guedes Barbosa Ramos
- Laboratory of Pharmacology and Molecular Chemistry, Department of Biological Chemistry, Regional University of Cariri, Rua Coronel Antônio Luis 1161, Crato 63105-000, CE, Brazil
| | - Maria Rayane Correia de Oliveira
- Laboratory of Pharmacology and Molecular Chemistry, Department of Biological Chemistry, Regional University of Cariri, Rua Coronel Antônio Luis 1161, Crato 63105-000, CE, Brazil
- Graduate Program in Biotechnology-Northeast Biotechnology Network (RENORBIO), State University of Ceará (UECE), Av. Dr. Silas Munguba, 1700, Fortaleza 60741-000, CE, Brazil
| | - Anita Oliveira Brito Pereira Bezerra Martins
- Laboratory of Pharmacology and Molecular Chemistry, Department of Biological Chemistry, Regional University of Cariri, Rua Coronel Antônio Luis 1161, Crato 63105-000, CE, Brazil
| | | | | | - Jaime Ribeiro-Filho
- Department of Biotechnology, Oswaldo Cruz Foundation, Fiocruz Ceará, Eusébio, Fortaleza 60180-190, CE, Brazil
| | | | - Henrique Douglas Melo Coutinho
- Laboratory of Microbiology and Molecular Biology, Department of Biological Chemistry, Regional University of Cariri, Rua Coronel Antônio Luis 1161, Crato 63105-000, CE, Brazil
| | - Irwin Rose Alencar de Menezes
- Laboratory of Pharmacology and Molecular Chemistry, Department of Biological Chemistry, Regional University of Cariri, Rua Coronel Antônio Luis 1161, Crato 63105-000, CE, Brazil
| |
Collapse
|
4
|
Pan W, Gu J, Xu S, Zhang C, Wang J, Wang S, Xu J. Dietary nitrate improves jaw bone remodelling in zoledronate-treated mice. Cell Prolif 2023:e13395. [PMID: 36810909 DOI: 10.1111/cpr.13395] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/16/2022] [Accepted: 12/21/2022] [Indexed: 02/24/2023] Open
Abstract
Bisphosphonate-related osteonecrosis of the jaw (BRONJ) is a serious complication that occurs in patients with osteoporosis or metastatic bone cancer treated with bisphosphonate. There is still no effective treatment and prevention strategy for BRONJ. Inorganic nitrate, which is abundant in green vegetables, has been reported to be protective in multiple diseases. To investigate the effects of dietary nitrate on BRONJ-like lesions in mice, we utilized a well-established mouse BRONJ model, in which tooth extraction was performed. Specifically, 4 mM sodium nitrate was administered in advance through drinking water to assess the short- and long-term effects on BRONJ. Zoledronate injection could induce severe healing inhibition of the tooth extraction socket, while addition of pretreating dietary nitrate could alleviate the inhibition by reducing monocyte necrosis and inflammatory cytokines production. Mechanistically, nitrate intake increased plasma nitric oxide levels, which attenuated necroptosis of monocytes by downregulating lipid and lipid-like molecule metabolism via a RIPK3 dependent pathway. Our findings revealed that dietary nitrate could inhibit monocyte necroptosis in BRONJ, regulate the bone immune microenvironment and promote bone remodelling after injury. This study contributes to the understanding of the immunopathogenesis of zoledronate and supports the feasibility of dietary nitrate for the clinical prevention of BRONJ.
Collapse
Affiliation(s)
- Wen Pan
- Salivary Gland Disease Centre and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jianyu Gu
- Salivary Gland Disease Centre and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Shihan Xu
- Salivary Gland Disease Centre and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chunmei Zhang
- Salivary Gland Disease Centre and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Immunology Research Centre for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Laboratory for Oral and General Health Integration and Translation, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jinsong Wang
- Salivary Gland Disease Centre and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Songlin Wang
- Salivary Gland Disease Centre and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Immunology Research Centre for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Laboratory for Oral and General Health Integration and Translation, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Research Units of Tooth Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Junji Xu
- Salivary Gland Disease Centre and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.,Immunology Research Centre for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Laboratory for Oral and General Health Integration and Translation, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Research Units of Tooth Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, China.,Department of Periodontics, Beijing Stomatological Hospital, Capital Medical University School of Stomatology, Beijing, China.,Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China
| |
Collapse
|
5
|
Protective Effect of Foxtail Millet Protein Hydrolysate on Ethanol and Pyloric Ligation-Induced Gastric Ulcers in Mice. Antioxidants (Basel) 2022; 11:antiox11122459. [PMID: 36552666 PMCID: PMC9774519 DOI: 10.3390/antiox11122459] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
Foxtail millet has been traditionally considered to possess gastroprotective effects, but studies evaluating its use as a treatment for gastric ulcers are lacking. Here, we assessed the antiulcer effects of foxtail millet protein hydrolysate (FPH) and explored its mechanism by using blocking agents. In a mouse model of ethanol-induced gastric ulcers, pretreatment with FPH reduced the ulcerative lesion index, downregulated the expression of inflammatory cytokines in the gastric tissue, increased the activity of antioxidant enzymes, and improved the oxidative status. FPH increased constitutive the activity of nitric oxide synthase (cNOS), NO levels, and mucin expression in gastric mucosa, and inhibited the activation of the ET-1/PI3K/Akt pathway. In a mouse model of pyloric ligation-induced gastric ulcers, FPH inhibited gastric acid secretion and decreased the activity of gastric protease. Pretreatment of mice with the sulfhydryl blocker NEM and the NO synthesis inhibitor L-NAME abolished the gastroprotective effect of FPH, but not the KATP channel blocker glibenclamide and the PGE2 synthesis blocker indomethacin. Among the peptides identified in FPH, 10 peptides were predicted to have regulatory effects on the gastric mucosa, and the key sequences were GP and PG. The results confirmed the gastroprotective effect of FPH and revealed that its mechanism was through the regulation of gastric mucosal mucus and NO synthesis. This study supports the health effects of a millet-enriched diet and provides a basis for millet protein as a functional food to improve gastric ulcers and its related oxidative stress.
Collapse
|
6
|
Alshora DH, Ibrahim MA, Zayed G, Al Rwashed MA, Abou-Taleb HA, Ali MF. The role of sodium lauryl sulfate on formulation of directly compressed tablets containing simvastatin and aspirin: Effect on drugs dissolution and gastric mucosa. Saudi Pharm J 2022; 30:635-645. [PMID: 35693440 PMCID: PMC9177453 DOI: 10.1016/j.jsps.2022.02.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 02/10/2022] [Indexed: 11/22/2022] Open
Abstract
According to the American College of Cardiology/American Heart Association (ACC/AHA), both aspirin and statin are used in the primary prevention of cardiovascular diseases. Aspirin (ASA) is contraindicated if there is gastrointestinal bleeding because it will exaggerate the condition. In this study, the effect of surfactant; sodium lauryl sulfate (SLS), in enhancing the in vitro dissolution of simvastatin (SIM) and ASA, as well as gastric irritation and upset, was studied. Oral tablets containing both ASA and SIM with and without the SLS were manufactured using the direct compression technique. The prepared tablets were characterized with respect to hardness, friability, uniformity of dosage units, in vitro disintegration, and dissolution. The effect of the addition of SLS in reducing the in vivo irritation and protection of gastric mucosa were also investigated. The results showed that the compressed tablets possessed sufficient hardness, acceptable friability, and are uniform with respect to disintegration, drugs contents, and tablet weight. The results showed that SIM alone exhibited a gastroprotective effect on the induced irritation. In addition, the incorporation of the SLS in the tablets containing SIM and ASA significantly enhanced the dissolution rates of both drugs and significantly decreased the gastric irritation and the ulcer index. The ulcer index of aspirin was decreased from 2.3 for tablets manufactured without SLS to 0.8 for tablets containing SLS. In a conclusion, the addition of pH modifier surfactant; SLS could enhance the dissolution rate of poorly soluble acidic drugs, reduce gastric upset and irritation without any effect on the main characters of the tablets. Moreover, the addition of SLS is very useful in improving the therapeutic activities and reducing the side effects of ASA and SIM for patients who require long-term administration of these drugs.
Collapse
Affiliation(s)
- Doaa H. Alshora
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Saudi Arabia
| | - Mohamed A. Ibrahim
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Gamal Zayed
- Department of Pharmaceutics and Industrial Pharmacy, Al-Azhar University, Assiut, Egypt
- Al-Azhar Centre of Nanosciences and Applications (ACNA), Al-Azhar University, Assiut, Egypt
| | | | - Heba A. Abou-Taleb
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Merit University (MUE), Sohag, Egypt
| | - Marwa F. Ali
- Department of Pathology and Clinical Pathology, Faculty of Veterinary Medicine, Assuit University, Egypt
| |
Collapse
|
7
|
Biano LS, Oliveira AS, Palmeira DN, Silva LA, de Albuquerque-Junior RLC, Duarte MC, Correa CB, Grespan R, Batista JS, Camargo EA. Gastroprotective action of the ethanol extract of Leonurus sibiricus L. (Lamiaceae) in mice. JOURNAL OF ETHNOPHARMACOLOGY 2022; 284:114792. [PMID: 34737011 DOI: 10.1016/j.jep.2021.114792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/14/2021] [Accepted: 10/27/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Leonurus sibiricus L. (Lamiaceae) is a medicinal plant known in Brazil as "rubim" or "erva de macaé". It is used for various purposes, including stomach disorders. AIM OF THE STUDY To evaluate the effect of the ethanol extract of the aerial parts of L. sibiricus (EELs) in models of gastric damage in mice. MATERIAL AND METHODS The effect of EELs (50, 100 and 300 mg/kg, p.o., 1 h before induction) was tested on acidified ethanol (ACEt)-induced gastric ulcers. Additionally, we tested the effect of EELs (by intraduodenal administration) in the pylorus ligation (PL) model. RESULTS Pretreatment with EELs, at 300 mg/kg, but not 50 and 100 mg/kg, reduced the relative area of gastric ulcers induced by ACEt (p < 0.01) and lipoperoxidation (p < 0.001), and increased the sulfhydryl content (p < 0.01) in the stomach in comparison with the vehicle group. Pretreatment with N-ethylmaleimide (a blocker of non-protein sulfhydryl groups, 10 mg/kg, i.p.) or glibenclamide (a KATP channel blocker, 10 mg/kg, i.p.) inhibited the gastroprotective response caused by EELs (300 mg/kg; p < 0.001), but there were no alterations due to pretreatments with inhibitors of the synthesis of prostaglandins (indomethacin, 10 mg/kg), nitric oxide (L-NAME, 70 mg/kg) or hydrogen sulfide (DL-propargylglycine, 10 mg/kg). Treatment with EELs (300 mg/kg) reduced mucus production (p < 0.001) and the volume of gastric secretion (p < 0.001) after PL without affecting gastric acidity or pH. CONCLUSIONS These results provide evidence that EELs exerts gastroprotective action in mice, with the participation of oxidative stress and mediation of NP-SH, KATP channels and mucus production.
Collapse
Affiliation(s)
- Laiza S Biano
- Graduation Program in Health Sciences, Federal University of Sergipe, Aracaju, SE, Brazil
| | - Alan S Oliveira
- Graduation Program in Health Sciences, Federal University of Sergipe, Aracaju, SE, Brazil
| | - David N Palmeira
- Department of Physiology, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | - Luis André Silva
- Graduation Program in Physiological Sciences, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | | | - Marcelo C Duarte
- Department of Pharmacy, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | - Cristiane B Correa
- Graduation Program in Health Sciences, Federal University of Sergipe, Aracaju, SE, Brazil; Graduation Program in Physiological Sciences, Federal University of Sergipe, São Cristóvão, SE, Brazil; Department of Morphology, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | - Renata Grespan
- Department of Physiology, Federal University of Sergipe, São Cristóvão, SE, Brazil; Graduation Program in Physiological Sciences, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | - Josemar S Batista
- Department of Physiology, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | - Enilton A Camargo
- Graduation Program in Health Sciences, Federal University of Sergipe, Aracaju, SE, Brazil; Department of Physiology, Federal University of Sergipe, São Cristóvão, SE, Brazil; Graduation Program in Physiological Sciences, Federal University of Sergipe, São Cristóvão, SE, Brazil.
| |
Collapse
|
8
|
Serafim CADL, Araruna MEC, Alves Júnior EB, Silva LMO, Silva AO, da Silva MS, Alves AF, Araújo AA, Batista LM. (-)-Carveol Prevents Gastric Ulcers via Cytoprotective, Antioxidant, Antisecretory and Immunoregulatory Mechanisms in Animal Models. Front Pharmacol 2021; 12:736829. [PMID: 34497525 PMCID: PMC8419343 DOI: 10.3389/fphar.2021.736829] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022] Open
Abstract
Background: (-)-Carveol (p-Mentha-6,8-dien-2-ol) is a monocyclic monoterpenic alcohol, present in essential oils of plant species such as Cymbopogon giganteus, Illicium pachyphyllum and in spices such as Carum carvi (cumin). Pharmacological studies report its antitumor, antimicrobial, neuroprotective, vasorelaxant, antioxidant and anti-inflammatory activity. Hypothesis/Purpose: The objective of this study was to evaluate the acute non-clinical oral toxicity, gastroprotective activity of monoterpene (-)-Carveol in animal models and the related mechanisms of action. Methods: Acute toxicity was assessed according to OECD guide 423 in mice. Ethanol, stress, NSAIDs and pylorus ligation-induced gastric ulcer models were used to investigate antiulcer properties. The related mechanisms of action were using the ethanol-gastric lesions protocol. Results: (-)-Carveol has low toxicity, with a lethal dose 50% (LD50) equal to or greater than 2,500 mg/kg according to OECD guide nº 423. In all gastric ulcer induction methods evaluated, (-)-Carveol (25, 50, 100 and 200 mg/kg, p.o.) significantly reduced the ulcerative lesion in comparison with the respective control groups. To investigate the mechanisms involved in the gastroprotective activity, the antisecretory or neutralizing of gastric secretion, cytoprotective, antioxidant and immunoregulatory effects were evaluated. In the experimental protocol of pylorus ligation-induced gastric ulcer, (-)-Carveol (100 mg/kg) reduced (p < 0.001) the volume of gastric secretion in both routes (oral and intraduodenal). The previous administration of blockers NEM (sulfhydryl groups blocker), L-NAME (nitric oxide synthesis inhibitor), glibenclamide (KATP channel blocker) and indomethacin (cyclo-oxygenase inhibitor), significantly reduced the gastroprotection exercised by (-)-Carveol, suggesting the participation of these pathways in its gastroprotective activity. In addition, treatment with (-)-Carveol (100 mg/kg) increased (p < 0.001) mucus adhered to the gastric wall. Treatment also increased (p < 0.001) levels of reduced glutathione (GSH), superoxide dismutase (SOD) and interleukin-10 (IL-10). It also reduced (p < 0.001) malondialdehyde (MDA), myeloperoxidase (MPO), interleukin-1 beta (IL-1β) and tumor necrosis factor-alpha (TNF-α) levels. Conclusion: Thus, it is possible to infer that (-)-Carveol presents gastroprotective activity related to antisecretory, cytoprotective, antioxidant and immunomodulatory mechanisms.
Collapse
Affiliation(s)
- Catarina Alves de Lima Serafim
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa, Brazil
| | - Maria Elaine Cristina Araruna
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa, Brazil
| | - Edvaldo Balbino Alves Júnior
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa, Brazil
| | - Leiliane Macena Oliveira Silva
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa, Brazil
| | - Alessa Oliveira Silva
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa, Brazil
| | - Marcelo Sobral da Silva
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa, Brazil
| | - Adriano Francisco Alves
- Department of Physiology and Pathology, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa, Brazil
| | - Aurigena Antunes Araújo
- Department of Morphology, Histology and Basic Pathology, Biosciences Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Leônia Maria Batista
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa, Brazil
| |
Collapse
|
9
|
Tamai R, Mashima I, Kiyoura Y. Alendronate Augments Lipid A-Induced IL-1α Release via Activation of ASC but Not Caspase-11. Inflammation 2021; 44:2132-2141. [PMID: 34080091 DOI: 10.1007/s10753-021-01489-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/10/2021] [Accepted: 05/24/2021] [Indexed: 11/26/2022]
Abstract
Nitrogen-containing bisphosphonates (NBPs), such as alendronate (ALN), are anti-bone-resorptive drugs that have inflammatory side effects. We previously reported that ALN augmented lipid A-induced interleukin (IL)-1β production and NOD-like receptor pyrin domain-containing-3 (NLRP3)/apoptosis-associated speck-like protein containing a CARD (ASC)-dependent cell death. The present study aimed to examine whether ALN augments lipid A-induced IL-1α release and necroptosis, which is induced by the activation of receptor-interacting protein kinase (RIPK) 3. Treatment of J774.1 cells with ALN augmented lipid A-induced IL-1α release, which was not inhibited by Ac-IETD-CHO, a caspase-8 inhibitor. ALN also activated mixed lineage kinase domain-like (MLKL), a key mediator of the necroptosis pathway, and upregulated the expression of caspase-11, a lipid A receptor. GSK'872, a RIPK3 inhibitor, suppressed the ALN-upregulated expression of caspase-11 and augmented lipid A-induced caspase-8 activation. Moreover, ALN induced the release of NLRP3 and ASC into culture supernatants. GSK'872, but not Ac-IETD-CHO, reduced the ALN-induced release of NLRP3, but not ASC, into culture supernatants, and reduced ALN-induced cell death, but not ALN-induced LDH release. Antibodies against NLRP3 and ASC upregulated caspase-11 expression in the cytosol by inhibiting ALN-induced cell death. However, pretreating cells with an antibody against ASC, but not NLRP3, before ALN addition also inhibited lipid A-induced IL-1α release. Pretreating cells with an antibody against caspase-11 before the addition of ALN or lipid A did not downregulate lipid A-induced production of IL-1α. Taken together, our findings suggest that ALN augments lipid A-induced IL-1α release via activation of ASC, but not caspase-11.
Collapse
Affiliation(s)
- Riyoko Tamai
- Department of Oral Medical Science, Ohu University School of Dentistry, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima, 963-8611, Japan.
| | - Izumi Mashima
- Department of Oral Medical Science, Ohu University School of Dentistry, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima, 963-8611, Japan
| | - Yusuke Kiyoura
- Department of Oral Medical Science, Ohu University School of Dentistry, 31-1 Misumido, Tomitamachi, Koriyama, Fukushima, 963-8611, Japan
| |
Collapse
|
10
|
Network Pharmacology Prediction and Pharmacological Verification Mechanism of Yeju Jiangya Decoction on Hypertension. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:5579129. [PMID: 34055010 PMCID: PMC8131144 DOI: 10.1155/2021/5579129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/13/2021] [Accepted: 04/28/2021] [Indexed: 02/06/2023]
Abstract
Background Yeju Jiangya decoction (CIF) is an herbal formula from traditional Chinese medicine (TCM) for the treatment of hypertension. Materials and Methods Based on the analysis of network pharmacology, combined with in animal experiments, the network pharmacology was used to explore the potential proteins and mechanisms of CIF against hypertension. The bioactive compounds of CIF were screened by using the platform, and the targets of hypertension and CIF were collected. Then, the Kyoto Encyclopedia of Genes and Genomes (KEGG) and protein-protein interaction network (PPI) core targets were carried out, and the useful proteins were found by molecular docking technology. Finally, we used N-nitro-L-arginine (L-NNA) induced hypertension model rats to confirm the effect and mechanism of CIF on hypertension. Results 14 bioactive compounds of CIF passed the virtual screening criteria, and 178 overlapping targets were identified as core targets of CIF against hypertension. The CIF-related target network with 178 nodes and 344 edges is constructed. The topological results show that quercetin and luteolin are the key components in the network. The key targets NOS3 (nitric oxide synthase 3) and NOS2 (nitric oxide synthase 2) were screened by the protein-protein interaction network. The analysis of target protein pathway enrichment showed that the accumulation pathway is related to the vascular structure of CIF regulation of hypertension. Further verification based on molecular docking results showed that NOS3 had the good binding ability with quercetin and luteolin. On the other hand, NOS3 has an important relationship with the composition of blood vessels. Furthermore, the animal experiment indicated that after the L-NNA-induced hypertension rat model was established, CIF intervention was given by gavage for 3 weeks, and it can decrease serum concentrations of endothelin-1 (ET-1) and thromboxane B2 (TXB2), increase the expression of nitric oxide (NO) and prostacyclin 2 (PGI2), and improve renal, cardiac, and aortic lesions. At the same time, it can reduce blood pressure and shorten vertigo time. Western blot (WB) and immunohistochemistry (IHC) analyses indicated that CIF may downregulate the expression of NOS3, guanylyl cyclase-alpha 1 (GC-α1), guanylyl cyclase-alpha 2 (GC-α2), and protein kinase CGMP-dependent 1 (PRKG1). These results suggest that CIF may play an antihypertensive role by inhibiting the activation of the NOS3/PRKG1 pathway. Conclusions The results of this study indicate that CIF has the ability to improve target organs, protect endothelial function, and reduce blood pressure and that CIF might be a potential therapeutic drug for the prevention of hypertension. It provides new insight into hypertension and the potential biological basis and mechanism for CIF clinical research.
Collapse
|
11
|
Pereira Júnior LC, Nascimento FG, Oliveira SRBD, Lima GC, Chagas FDS, Sombra VG, Feitosa JPA, Soriano EM, Souza MHLP, Zocolo GJ, Silva LMA, de Paula RCM, Damasceno ROS, Freitas ALP. Protective effect against gastric mucosa injury of a sulfated agaran from Acanthophora spicifera. Carbohydr Polym 2021; 261:117829. [PMID: 33766334 DOI: 10.1016/j.carbpol.2021.117829] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 02/09/2021] [Accepted: 02/12/2021] [Indexed: 11/26/2022]
Abstract
In this study, a polysaccharide from marine alga Acanthophora spicifera (PAs) was isolated and structurally characterized. Its protective potential against chemically-induced gastric mucosa injury was evaluated. The gel permeation chromatography experiments and spectroscopy spectrum showed that PAs is a sulfated polysaccharide with a high molecular mass (6.98 × 105g/mol) and degree of sulfation of 1.23, exhibiting structural characteristic typical of an agar-type polysaccharide. Experimental results demonstrated that PAs reduced the hemorrhagic gastric injury, in a dose-dependent manner. Additionally, PAs reduced the intense gastric oxidative stress, measured by glutathione (GSH) and malondialdehyde (MDA) levels. PAs also prevented the reduction of mucus levels adhered to the gastric mucosa, promoted by the aggressive effect of ethanol. In summary, the sulfated polysaccharide from A. spicifera protected the gastric mucosa through the prevention of lipid peroxidation and enhanced the defense mechanisms of the gastric mucosa, suggesting as a promising functional food as gastroprotective agent.
Collapse
Affiliation(s)
- Lindauro C Pereira Júnior
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Ceará, 60455-760, Fortaleza, CE, Brazil
| | | | - Samara R B D Oliveira
- Departamento de Fisiologia e Farmacologia, Universidade Federal do Ceará, 60430-270, Fortaleza, CE, Brazil
| | - Glauber C Lima
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Ceará, 60455-760, Fortaleza, CE, Brazil; Centro Universitário INTA (UNINTA), 62500-000, Itapipoca, CE, Brazil
| | - Francisco Diego S Chagas
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Ceará, 60455-760, Fortaleza, CE, Brazil
| | - Venicios G Sombra
- Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, 60455-760, Fortaleza, CE, Brazil
| | - Judith P A Feitosa
- Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, 60455-760, Fortaleza, CE, Brazil
| | - Eliane M Soriano
- Departamento de Oceanografia e Limnologia, Universidade Federal do Rio Grande do Norte, 59072-970, Natal, RN, Brazil
| | - Marcellus H L P Souza
- Departamento de Fisiologia e Farmacologia, Universidade Federal do Ceará, 60430-270, Fortaleza, CE, Brazil
| | | | - Lorena M A Silva
- Embrapa Agroindústria Tropical, 60511-110, Fortaleza, CE, Brazil
| | - Regina C M de Paula
- Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, 60455-760, Fortaleza, CE, Brazil
| | - Renan O S Damasceno
- Departamento de Fisiologia e Farmacologia, Universidade Federal de Pernambuco, 50670-420, Recife, PE, Brazil.
| | - Ana Lúcia P Freitas
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Ceará, 60455-760, Fortaleza, CE, Brazil
| |
Collapse
|
12
|
MPMBP down-regulates Toll-like receptor (TLR) 2 ligand-induced proinflammatory cytokine production by inhibiting NF-κB but not AP-1 activation. Int Immunopharmacol 2020; 79:106085. [DOI: 10.1016/j.intimp.2019.106085] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 11/23/2019] [Accepted: 11/25/2019] [Indexed: 12/21/2022]
|
13
|
Magierowska K, Korbut E, Hubalewska-Mazgaj M, Surmiak M, Chmura A, Bakalarz D, Buszewicz G, Wójcik D, Śliwowski Z, Ginter G, Gromowski T, Kwiecień S, Brzozowski T, Magierowski M. Oxidative gastric mucosal damage induced by ischemia/reperfusion and the mechanisms of its prevention by carbon monoxide-releasing tricarbonyldichlororuthenium (II) dimer. Free Radic Biol Med 2019; 145:198-208. [PMID: 31568823 DOI: 10.1016/j.freeradbiomed.2019.09.032] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/25/2019] [Accepted: 09/26/2019] [Indexed: 02/08/2023]
Abstract
Endogenous gaseous mediators, such as nitric oxide, hydrogen sulfide or carbon monoxide (CO) are known to exert anti-inflammatory and anti-oxidative activity due to modulation of various molecular pahtways. Therefore, we aimed to investigate if CO released from tricarbonyldichlororuthenium (II) dimer (CORM-2) prevents gastric mucosa against ischemia/reperfusion (I/R)-induced injury in male Wistar rats. Animals were pretreated i.g. With vehicle (DMSO and saline, 1:10), CORM-2 (1, 5 or 10 mg/kg) or zinc protoporphyrin IX (ZnPP, 10 mg/kg i.p.), the HMOXs inhibitor. In separate series, rats were pretreated with CORM-2 (5 mg/kg) applied in combination with glibenclamide (10 mg/kg i.g.), NG-nitro-l-arginine (L-NNA, 20 mg/kg i.p.), 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ, 10 mg/kg i.p.) or indomethacin (5 mg/kg i.p.). I/R-injuries were induced by clamping celiac artery for 30 min (I) followed by removal of the clamp to obtain R for 3 h. The macroscopic and microscopic area of gastric damage, mucus production and protein expression for HMOX-1/Nrf-2 was determined by planimetry, histology and immunohistochemistry, respectively. Gastric mucosal HMOX-1, HMOX-2, COX-1, COX-2, Kir6.1, Sur2, sGC-α1, sGC-α2, iNOS and eNOS mRNA expression was assessed by real-time PCR. COHb in blood and gastric mucosal CO concentration was analyzed by gas chromatography. Serum content of TGF-β1, TGF-β2, TGF-β3, IL-1α, IL-1β, IL-2, IL-4, IL-5, IL-6, IL-10, IL-12, TNF-α, IFN-γ, GM-CSF was evaluated using Luminex platform. PGE2 concentration and 8-hydroxyguanozine (8-OHG) concentration in gastric mucosa was determined by ELISA. Exposure to I/R induced extensive hemorrhagic erosions in gastric mucosa pretreated with vehicle as compared with intact rats and the area of this gastric damage was reduced by pretreatment with CORM-2 (5 mg/kg i.g.). This effect of CO donor was accompanied by the increased PGE2 content and a significant decrease in 8-OHG and expression of pro- and anti-inflammatory markers mRNA and proteins. Concurrent treatment of CORM-2 with glibenclamide, L-NNA, ODQ but not with indomethacin significantly increased the area of I/R-induced injury and significantly decreased GBF as compared with the group treated with CORM-2 alone. We conclude that CO releasing CORM-2 prevents gastric mucosal oxidative damage induced by I/R improving GBF, decreasing DNA oxidation and inflammatory response on systemic level. This CO-gastroprotection is mediated by the activity of sGC, NOS and K-ATP channels. CO delivered from its donor maintained physiological gastric mucosal PGE2 concentration but the involvement of endogenous COX in beneficial activity of this gaseous mediator at least in this model is questionable.
Collapse
Affiliation(s)
| | - Edyta Korbut
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | | | - Marcin Surmiak
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | - Anna Chmura
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | - Dominik Bakalarz
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland; Department of Forensic Toxicology, Institute of Forensic Research, Cracow, Poland
| | - Grzegorz Buszewicz
- Department of Forensic Medicine, Medical University of Lublin, Lublin, Poland
| | - Dagmara Wójcik
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | - Zbigniew Śliwowski
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | - Grzegorz Ginter
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | - Tomasz Gromowski
- Human Genome Variation Research Group & Genomics Centre, Malopolska Centre of Biotechnology, Jagiellonian University, Cracow, Poland
| | - Sławomir Kwiecień
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | - Tomasz Brzozowski
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | - Marcin Magierowski
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland.
| |
Collapse
|
14
|
Nolêto IR, Iles B, Alencar MS, Lopes AL, Oliveira AP, Pacheco G, Sousa FB, Araújo AR, Alves EH, Vasconcelos DF, Leal LKA, Araújo AJ, Filho JDB, Medeiros JVR. Alendronate-induced gastric damage in normoglycemic and hyperglycemic rats is reversed by metformin. Eur J Pharmacol 2019; 856:172410. [DOI: 10.1016/j.ejphar.2019.172410] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/17/2019] [Accepted: 05/22/2019] [Indexed: 02/07/2023]
|
15
|
Pócs L, Janovszky Á, Ocsovszki I, Kaszaki J, Piffkó J, Szabó A. Microcirculatory consequences of limb ischemia/reperfusion in ovariectomized rats treated with zoledronic acid. J Orthop Surg Res 2019; 14:95. [PMID: 30947735 PMCID: PMC6450009 DOI: 10.1186/s13018-019-1117-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 03/06/2019] [Indexed: 12/12/2022] Open
Abstract
Background Nitrogen-containing bisphosphonates (BIS) are potent therapeutics in osteoporosis, but their use may result in osteonecrotic side-effects in the maxillofacial region. Periosteal microcirculatory reactions may contribute to the development of bone-healing complications, particularly in osteoporotic bones, where ischemia–reperfusion (IR) events often develop during orthopaedic/trauma interventions. The effect of BIS on the inflammatory reactions of appendicular long bones has not yet been evaluated; thus, we aimed to examine the influence of chronic zoledronate (ZOL) administration on the periosteal microcirculatory consequences of hindlimb IR in osteopenic rats. Materials and methods Twelve-week-old female Sprague–Dawley rats were ovariectomized (OVX) or sham-operated, and ZOL (80 μg/kg iv, weekly) or a vehicle was administered for 8 weeks, 4 weeks after the operation. At the end of the pre-treatment protocols, 60-min limb ischemia was induced, followed by 180-min reperfusion. Leukocyte-endothelial interactions were quantitated in tibial periosteal postcapillary venules by intravital fluorescence videomicroscopy. CD11b expression of circulating polymorphonuclear leukocytes (PMN, flow cytometry) and plasma TNF-alpha levels (ELISA) were also determined. Two-way RM ANOVA followed by the Holm–Sidak and Dunn tests was used to assess differences within and between groups, respectively. Results Limb IR induced significant increases in PMN rolling and firm adhesion in sham-operated and OVX rats, which were exacerbated temporarily in the first 60 min of reperfusion by a ZOL treatment regimen. Postischemic TNF-alpha values showed a similar level of postischemic elevations in all groups, whereas CD11b expression only increased in rats not treated with ZOL. Conclusions The present data do not show substantial postischemic periosteal microcirculatory complications after chronic ZOL treatment either in sham-operated or OVX rats. The unaltered extent of limb IR-induced local periosteal microcirculatory reactions in the presence of reduced CD11b adhesion molecule expression on circulating PMNs, however, may be attributable to local endothelial injury/activation caused by ZOL.
Collapse
Affiliation(s)
- Levente Pócs
- Department of Traumatology and Hand Surgery, Bács-Kiskun County Teaching Hospital, Nyíri u. 38, Kecskemét, H-6000, Hungary
| | - Ágnes Janovszky
- Department of Oral and Maxillofacial Surgery, University of Szeged, Kálvária sgt. 57, Szeged, H-6725, Hungary
| | - Imre Ocsovszki
- Department of Biochemistry, University of Szeged, Dóm tér 9, Szeged, H-6720, Hungary
| | - József Kaszaki
- Department of Oral and Maxillofacial Surgery, University of Szeged, Kálvária sgt. 57, Szeged, H-6725, Hungary
| | - József Piffkó
- Department of Oral and Maxillofacial Surgery, University of Szeged, Kálvária sgt. 57, Szeged, H-6725, Hungary
| | - Andrea Szabó
- Institute of Surgical Research, University of Szeged, Pulz u. 1, Szeged, H-6724, Hungary.
| |
Collapse
|
16
|
Chiesa JJ, Baidanoff FM, Golombek DA. Don't just say no: Differential pathways and pharmacological responses to diverse nitric oxide donors. Biochem Pharmacol 2018; 156:1-9. [PMID: 30080991 DOI: 10.1016/j.bcp.2018.08.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/02/2018] [Indexed: 12/18/2022]
Abstract
Nitric oxide (NO) is a gaseous free radical molecule with a short half-life (∼1 s), which can gain or lose an electron into three interchangeable redox-dependent forms, the radical (NO), the nitrosonium cation (NO+), and nitroxyl anion (HNO). NO acts as an intra and extracellular signaling molecule regulating a wide range of functions in the cardiovascular, immune, and nervous system. NO donors are collectively known by their ability to release NOin vitro and in vivo, being proposed as therapeutic pharmacological tools for the treatment of several pathologies, such as cardiovascular disease. The highly reactive NO molecule is easily oxidized under physiological conditions to N-oxides, nitrate/nitrite and nitrogen dioxide. Different cellular responses are triggered depending on: 1) NO concentration [e.g., nanomolar for heme coordination in the allosteric site of guanylate cyclase (sGC) enzyme]; 2) the type of chemical bound to the nitrosated group (i.e., bound to nitrogen, N-nitro, or bound to sulphur atom, S-nitro) determining post-translational cysteine nitrosation; 3) the time-dependent availability of molecular targets. Classic NO donors are: organic nitrates (e.g., nitroglycerin, or glyceryl trinitrate, GTN; isosorbide mononitrate, ISMN), diazeniumdiolates having a diolate group [or NONOates, e.g., 2-(N,N-diethylamino)-diazenolate-2-oxide], S-nitrosothiols (e.g., S-nitroso glutathione, GSNO; S-nitroso-N-acetylpenicillamine, SNAP) or the organic salt sodium nitroprusside (SNP). In addition, nitroxyl (HNO) donors such as Piloty's acid and Angeli's salt can also be considered. The specific NO form released, as well as its differential reactivity to thiols, could act on different molecular targets and should be discussed in the context of: a) the type and amount of NO species determining the sensitivity of molecular targets (e.g., heme coordination, or S-nitrosation); b) the cellular redox state that could gate different effects. Experimental designs should take special care when choosing which NO donors to use, since different outcomes are to be expected. This article will comment recent findings regarding physiological responses involving NO species and their pharmacological modulation with donor drugs, especially in the context of the photic transduction pathways at the hypothalamic circadian clock.
Collapse
Affiliation(s)
- Juan J Chiesa
- Laboratorio de Cronobiología, Universidad Nacional de Quilmes/CONICET, Argentina
| | - Fernando M Baidanoff
- Laboratorio de Cronobiología, Universidad Nacional de Quilmes/CONICET, Argentina
| | - Diego A Golombek
- Laboratorio de Cronobiología, Universidad Nacional de Quilmes/CONICET, Argentina.
| |
Collapse
|
17
|
de Araújo S, Oliveira AP, Sousa FBM, Souza LKM, Pacheco G, Filgueiras MC, Nicolau LAD, Brito GAC, Cerqueira GS, Silva RO, Souza MHLP, Medeiros JVR. AMPK activation promotes gastroprotection through mutual interaction with the gaseous mediators H 2S, NO, and CO. Nitric Oxide 2018; 78:60-71. [PMID: 29857061 DOI: 10.1016/j.niox.2018.05.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 04/25/2018] [Accepted: 05/29/2018] [Indexed: 12/17/2022]
Abstract
Activation of 5' adenosine monophosphate-activated protein kinase (AMPK) stimulates production of the gaseous mediators nitric oxide (NO) and carbon monoxide (CO), which are involved in mucosal defense and gastroprotection. As AMPK itself has gastroprotective effects against several gastric ulcer etiologies, in the present study, we aimed to elucidate whether AMPK may also prevent ethanol-induced injury and play a key role in the associated gastroprotection mediated by hydrogen sulfide (H2S), NO, and CO. Mice were pretreated with AICAR (20 mg/kg, an AMPK activator) alone or with 50% ethanol. Other groups were pretreated with respective gaseous mediator inhibitors PAG, l-NAME, or ZnPP IX 30 min prior to AICAR, or with gaseous mediator donors NaHS, Lawesson's reagent and l-cysteine (H2S), SNP, l-Arginine (NO), Hemin, or CORM-2 (CO) 30 min prior to ethanol with or without compound C (10 mg/kg, a non-selective AMPK inhibitor). H2S, nitrate/nitrite (NO3-/NO2-), bilirubin levels, GSH and MDA concentration were evaluated in the gastric mucosa. The gastric mucosa was also collected for histopathological analysis and AMPK expression assessment by immunohistochemistry. Pretreatment with AICAR attenuated the ethanol-induced injury and increased H2S and bilirubin levels but not NO3-/NO2- levels in the gastric mucosa. In addition, inhibition of H2S, NO, or CO synthesis exacerbated the ethanol-induced gastric damage and inhibited the gastroprotection by AICAR. Pretreatment with compound C reversed the gastroprotective effect of NaHS, Lawesson's reagent, l-cysteine, SNP, l-Arginine, CORM-2, or Hemin. Compound C also reversed the effect of NaHS on H2S production, SNP on NO3-/NO2- levels, and Hemin on bilirubin levels. Immunohistochemistry revealed that AMPK is present at basal levels mainly in the gastric mucosa cells, and was increased by pretreatment with NaHS, SNP, and CORM-2. In conclusion, our findings indicate that AMPK activation exerts gastroprotection against ethanol-induced gastric damage and mutually interacts with H2S, NO, or CO to facilitate this process.
Collapse
Affiliation(s)
- Simone de Araújo
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Federal University of Piauí, Parnaíba, Piauí, Brazil
| | - Ana P Oliveira
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Federal University of Piauí, Parnaíba, Piauí, Brazil
| | - Francisca B M Sousa
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Federal University of Piauí, Parnaíba, Piauí, Brazil
| | - Luan K M Souza
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Federal University of Piauí, Parnaíba, Piauí, Brazil
| | - Gabriella Pacheco
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Federal University of Piauí, Parnaíba, Piauí, Brazil
| | - Marcelo C Filgueiras
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Federal University of Piauí, Parnaíba, Piauí, Brazil
| | - Lucas A D Nicolau
- Departments of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Gerly Anne C Brito
- Postgraduate Program in Morphofunctional Sciences, Department of Morphology, Faculty of Medicine, Federal University Ceará, Fortaleza, Ceará, Brazil
| | - Gilberto S Cerqueira
- Postgraduate Program in Morphofunctional Sciences, Department of Morphology, Faculty of Medicine, Federal University Ceará, Fortaleza, Ceará, Brazil
| | - Renan O Silva
- Departments of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Marcellus H L P Souza
- Departments of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Jand Venes R Medeiros
- Laboratory of Pharmacology of Inflammation and Gastrointestinal Disorders (LAFIDG), Federal University of Piauí, Parnaíba, Piauí, Brazil.
| |
Collapse
|
18
|
Tamai R, Kiyoura Y. Alendronate augments lipid A-induced IL-1β release and Smad3/NLRP3/ASC-dependent cell death. Life Sci 2018; 198:8-17. [DOI: 10.1016/j.lfs.2018.02.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 02/01/2018] [Accepted: 02/09/2018] [Indexed: 12/13/2022]
|
19
|
Magierowski M, Magierowska K, Hubalewska‐Mazgaj M, Sliwowski Z, Ginter G, Pajdo R, Chmura A, Kwiecien S, Brzozowski T. Carbon monoxide released from its pharmacological donor, tricarbonyldichlororuthenium (II) dimer, accelerates the healing of pre-existing gastric ulcers. Br J Pharmacol 2017; 174:3654-3668. [PMID: 28768046 PMCID: PMC5610153 DOI: 10.1111/bph.13968] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 07/10/2017] [Accepted: 07/25/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Carbon monoxide (CO), a gaseous mediator produced by haem oxygenases (HOs), has been shown to prevent stress-, ethanol-, aspirin- and alendronate-induced gastric damage; however, its role in gastric ulcer healing has not been fully elucidated. We investigated whether CO released from tricarbonyldichlororuthenium (II) dimer (CORM-2) can affect gastric ulcer healing and determined the mechanisms involved in this healing action. EXPERIMENTAL APPROACH Gastric ulcers were induced in Wistar rats by serosal application of acetic acid. Animals received 9 days of treatment with RuCl3 [2.5 mg·kg-1 intragastrically (i.g.)], haemin (5 mg·kg-1 i.g.), CORM-2 (0.1-10 mg·kg-1 i.g.) administered alone or with zinc protoporphyrin IX (ZnPP, 10 mg·kg-1 i.g.), 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ, 5 mg·kg-1 i.g.), NG -nitro-l-arginine (l-NNA, 15 mg·kg-1 i.g.), indomethacin (5 mg·kg-1 i.g.) or glibenclamide (10 mg·kg-1 i.g.). Gastric ulcer area and gastric blood flow (GBF) were assessed planimetrically, microscopically and by laser flowmeter respectively. Gastric mRNA/protein expressions of EGF, EGF receptors, VEGFA, HOs, nuclear factor (erythroid-derived 2)-like 2 (Nrf2), COX-2, hypoxia-inducible factor (HIF)-1α and pro-inflammatory iNOS, IL-1β and TNF-α were determined by real-time PCR or Western blots. KEY RESULTS CORM-2 and haemin but not RuCl3 or ZnPP decreased ulcer size while increasing GBF. These effects were reduced by ODQ, indomethacin, l-NNA and glibenclamide. CORM-2 significantly decreased the expression of pro-inflammatory markers, Nrf2/HO1 and HIF-1α, and up-regulated EGF. CONCLUSIONS AND IMPLICATIONS CO released from CORM-2 or endogenously produced by the HO1/Nrf2 pathway accelerates gastric ulcer healing via an increase in GBF, an up-regulation in EGF expression and down-regulation of the inflammatory response.
Collapse
Affiliation(s)
- Marcin Magierowski
- Department of Physiology, Faculty of MedicineJagiellonian University Medical CollegeCracowPoland
| | - Katarzyna Magierowska
- Department of Physiology, Faculty of MedicineJagiellonian University Medical CollegeCracowPoland
| | | | - Zbigniew Sliwowski
- Department of Physiology, Faculty of MedicineJagiellonian University Medical CollegeCracowPoland
| | - Grzegorz Ginter
- Department of Physiology, Faculty of MedicineJagiellonian University Medical CollegeCracowPoland
| | - Robert Pajdo
- Department of Physiology, Faculty of MedicineJagiellonian University Medical CollegeCracowPoland
| | - Anna Chmura
- Department of Physiology, Faculty of MedicineJagiellonian University Medical CollegeCracowPoland
| | - Slawomir Kwiecien
- Department of Physiology, Faculty of MedicineJagiellonian University Medical CollegeCracowPoland
| | - Tomasz Brzozowski
- Department of Physiology, Faculty of MedicineJagiellonian University Medical CollegeCracowPoland
| |
Collapse
|
20
|
Zazueta-Beltrán L, Medina-Aymerich L, Estela Díaz-Triste N, Chávez-Piña AE, Castañeda-Hernández G, Cruz-Antonio L. Evidence against the participation of a pharmacokinetic interaction in the protective effect of single-dose curcumin against gastrointestinal damage induced by indomethacin in rats. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2017; 15:151-157. [PMID: 28285620 DOI: 10.1016/s2095-4964(17)60324-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To determine the role of a pharmacokinetic interaction in the protective effect of curcumin against the gastric damage induced by indomethacin administration as such or as its prodrug acemetacin. METHODS Wistar rats orally received single dose of indomethacin (30 mg/kg) with and without curcumin (30 mg/kg); gastric injury was evaluated by determining the total damaged area. Additional groups of rats received an oral single dose of indomethacin (30 mg/kg) or its prodrug acemetacin (34.86 mg/kg) in the presence or absence of curcumin (30 mg/kg). Indomethacin and acemetacin concentrations in plasma from blood draws were determined by high-performance liquid chromatography.Plasma concentration-against-time curves were constructed, and bioavailability parameters, maximal concentration (Cmax) and area under the curve to the last sampling time (AUC0-t) were estimated. RESULTS Concomitant administration of indomethacin and curcumin resulted in a significantly reduced gastric damage compared to indomethacin alone. However, co-administration of curcumin did not produce any significant alteration in the bioavailability parameters of indomethacin and acemetacin after administration of either the active compound or the prodrug. CONCLUSION Curcumin exhibits a protective effect against indomethacin-induced gastric damage, but does not produce a reduction of the bioavailability of this nonsteroidal anti-inflammatory drug, indomethacin. Data thus suggest that a pharmacokinetic mechanism of action is not involved in curcumin gastroprotection.
Collapse
Affiliation(s)
- Liliana Zazueta-Beltrán
- Department of Pharmacology, Center for Research and Advanced Studies of the National Polytechnic Institute, 07360 Mexico City, Mexico
| | - Lorena Medina-Aymerich
- Department of Pharmacology, Center for Research and Advanced Studies of the National Polytechnic Institute, 07360 Mexico City, Mexico
| | - Nadia Estela Díaz-Triste
- Department of Pharmacology, Center for Research and Advanced Studies of the National Polytechnic Institute, 07360 Mexico City, Mexico
| | - Aracely Evangelina Chávez-Piña
- Laboratory of Pharmacology, Institutional Program in Molecular Biomedicine, National School of Medicine and Homeopathy of the National Polytechnic Institute, Academic Area of Medicine of the Institute of Health Sciences, 07320 Mexico City, Mexico
| | - Gilberto Castañeda-Hernández
- Department of Pharmacology, Center for Research and Advanced Studies of the National Polytechnic Institute, 07360 Mexico City, Mexico
| | - Leticia Cruz-Antonio
- Superior Studies Faculty of Zaragoza, Universidad National Autonomous of Mexico, 09230 Mexico City, Mexico
| |
Collapse
|
21
|
Nitric Oxide and Hydrogen Sulfide Interact When Modulating Gastric Physiological Functions in Rodents. Dig Dis Sci 2017; 62:93-104. [PMID: 27864656 DOI: 10.1007/s10620-016-4377-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 11/07/2016] [Indexed: 12/13/2022]
Abstract
AIM The objective was to evaluate the effects of nitric oxide (NO) and hydrogen sulfide (H2S) donors and possible interactions between these two systems in modulating gastric function. METHODS Mice received saline, sodium nitroprusside (SNP), or sodium hydrosulfite (NaHS), and after 1 h, the animals were killed for immunofluorescence analysis of CSE or eNOS expressions, respectively. Other groups received saline, SNP, NaHS, Lawesson's reagent (H2S donor), PAG + SNP, L-NAME, L-NAME + NaHS, or L-NAME + Lawesson's reagent. Then, the gastric secretions (mucous and acid), gastric blood flow, gastric defense against ethanol, and gastric motility (gastric emptying and gastric contractility) were evaluated. RESULTS SNP and NaHS increased the expression of CSE or eNOS, respectively. SNP or Lawesson's reagent did not alter gastric acid secretion but increased mucus production, and these effects reverted with PAG and L-NAME treatment, respectively. SNP or NaHS increased gastric blood flow and protected the gastric mucosa against ethanol injury, and these effects reverted with PAG and L-NAME treatments, respectively. SNP delayed gastric emptying when compared with saline, and PAG partially reversed this effect. NaHS accelerate gastric emptying, and L-NAME partially reversed this effect. SNP and NaHS alone induced gastric fundus and pylorus relaxation. However, pretreatment with PAG or L-NAME reversed these relaxant effects only in the pylorus but not in the gastric fundus. CONCLUSION NO and H2S interact in gastric physiological functions, and this "cross-talk" is important in the control of mucus secretion, gastric blood flow, gastric mucosal defense, and gastric motility, but not in the control of basal gastric acid secretion.
Collapse
|
22
|
Magierowski M, Magierowska K, Szmyd J, Surmiak M, Sliwowski Z, Kwiecien S, Brzozowski T. Hydrogen Sulfide and Carbon Monoxide Protect Gastric Mucosa Compromised by Mild Stress Against Alendronate Injury. Dig Dis Sci 2016; 61:3176-3189. [PMID: 27541924 PMCID: PMC5067292 DOI: 10.1007/s10620-016-4280-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 08/10/2016] [Indexed: 01/08/2023]
Abstract
BACKGROUND Alendronate is an inhibitor of osteoclast-mediated bone resorption, but its clinical utility is limited due to gastrointestinal complications including bleeding erosions. AIMS We studied whether potent vasodilators hydrogen sulfide (H2S) and carbon monoxide (CO) can protect against alendronate-induced gastric lesions in rats exposed to mild stress. METHODS Three series (A, B, and C) of Wistar rats received alendronate (150-700 mg/kg i.g., series A) with or without NaHS (5 mg/kg), H2S donor or CORM-2 (5 mg/kg) releasing CO administered i.g. 30 min before alendronate administration (series B) in rats exposed for 3 days before alendronate administration to mild stress (series C). The area of gastric lesions was assessed by planimetry, the gastric blood flow (GBF) was determined by H2-gas clearance technique, and H2S production via CSE/CBS/3-MST activity and the gastric expression of HO-1, HO-2, HIF-1α, NF-κB, iNOS, COX-2, IL-1β, TNF-α, GPx-1 and SOD-2 were analyzed by qPCR or Western blot. RESULTS Alendronate dose-dependently produced gastric mucosal lesions and significantly decreased GBF, and these effects were exacerbated by mild stress. NaHS and CORM-2 significantly reduced the alendronate-induced gastric lesions in non-stressed and stressed animals, but only NaHS but not CORM-2 raised H2S production. NaHS and CORM-2 inhibited gastric expression of HIF-1α protein and HO-1, HIF-1α, NF-κB, COX-2, iNOS, IL-1β, TNF-α mRNAs but failed to affect those of HO-2, GPx-1, and SOD-2. CONCLUSION Both H2S and CO released from their donors, NaHS and CORM-2, protect gastric mucosa compromised by stress against alendronate-induced gastric damage via mechanism involving downregulation of HIF-1α, NF-κB and proinflammatory factors COX-2, iNOS, IL-1β, and TNF-α.
Collapse
Affiliation(s)
- Marcin Magierowski
- Department of Physiology, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Cracow, Poland
| | - Katarzyna Magierowska
- Department of Physiology, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Cracow, Poland
| | - Jakub Szmyd
- Department of Physiology, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Cracow, Poland
| | - Marcin Surmiak
- Department of Physiology, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Cracow, Poland
- Division of Molecular Biology and Clinical Genetics, Department of Medicine, Jagiellonian University Medical College, 8 Skawinska Street, 31-066 Cracow, Poland
| | - Zbigniew Sliwowski
- Department of Physiology, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Cracow, Poland
| | - Slawomir Kwiecien
- Department of Physiology, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Cracow, Poland
| | - Tomasz Brzozowski
- Department of Physiology, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531 Cracow, Poland
| |
Collapse
|
23
|
Cutini PH, Rauschemberger MB, Sandoval MJ, Massheimer VL. Vascular action of bisphosphonates: In vitro effect of alendronate on the regulation of cellular events involved in vessel pathogenesis. J Mol Cell Cardiol 2016; 100:83-92. [PMID: 27705747 DOI: 10.1016/j.yjmcc.2016.08.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 08/19/2016] [Accepted: 08/24/2016] [Indexed: 11/28/2022]
Abstract
In this work we investigate whether, despite the procalcific action of alendronate on bone, the drug would be able to regulate in vitro the main cellular events that take part in atherosclerotic lesion generation. Using endothelial cell cultures we showed that Alendronate (1-50μM) acutely enhances nitric oxide production (10-30min). This stimulatory action of the bisphosphonate involves the participation of MAPK signaling transduction pathway. Under inflammatory stress, the drug reduces monocytes and platelets interactions with endothelial cells induced by lipopolysaccharide. Indeed the bisphophonate exhibits a significant inhibition of endothelial dependent platelet aggregation. The molecular mechanism of alendronate (ALN) on leukocyte adhesion depends on the regulation of the expression of cell adhesion related genes (VCAM-1; ICAM-1); meanwhile the antiplatelet activity is associated with the effect of the drug on nitric oxide production. On vascular smooth muscle cells, the drug exhibits ability to decrease osteogenic transdifferentiation and extracellular matrix mineralization. When vascular smooth muscle cells were cultured in osteogenic medium for 21days, they exhibited an upregulation of calcification markers (RUNX2 and TNAP), high alkaline phosphatase activity and a great amount of mineralization nodules. ALN treatment significantly down-regulates mRNA levels of osteoblasts markers; diminishes alkaline phosphatase activity and reduces the extracellular calcium deposition. The effect of ALN on vascular cells differs from its own bone action. On calvarial osteoblasts ALN induces cell proliferation, enhances alkaline phosphatase activity, and increases mineralization, but does not affect nitric oxide synthesis. Our results support the hypothesis that ALN is an active drug at vascular level that regulates key processes involved in vascular pathogenesis through a direct action on vessel cells.
Collapse
Affiliation(s)
- Pablo H Cutini
- Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR), Universidad Nacional del Sur (UNS), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Departamento de Biología, Bioquímica y Farmacia, San Juan 670, B8000ICN, Bahía Blanca, Argentina.
| | - María B Rauschemberger
- Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR), Universidad Nacional del Sur (UNS), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Departamento de Biología, Bioquímica y Farmacia, San Juan 670, B8000ICN, Bahía Blanca, Argentina.
| | - Marisa J Sandoval
- Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR), Universidad Nacional del Sur (UNS), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Departamento de Biología, Bioquímica y Farmacia, San Juan 670, B8000ICN, Bahía Blanca, Argentina.
| | - Virginia L Massheimer
- Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR), Universidad Nacional del Sur (UNS), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Departamento de Biología, Bioquímica y Farmacia, San Juan 670, B8000ICN, Bahía Blanca, Argentina.
| |
Collapse
|
24
|
Zheng H, Chen Y, Zhang J, Wang L, Jin Z, Huang H, Man S, Gao W. Evaluation of protective effects of costunolide and dehydrocostuslactone on ethanol-induced gastric ulcer in mice based on multi-pathway regulation. Chem Biol Interact 2016; 250:68-77. [PMID: 26970604 DOI: 10.1016/j.cbi.2016.03.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Revised: 02/22/2016] [Accepted: 03/01/2016] [Indexed: 10/22/2022]
Abstract
The aim of the present study was to evaluate the anti-ulcerogenic activity of costunolide (Co) and dehydrocostuslactone (De) on ethanol-induced gastric ulcer in mice and to elucidate the potential mechanisms of the action involved. Mice were pretreated orally with Co (5 or 20 mg/kg), De (5 or 20 mg/kg) and omeprazole (OME, 20 mg/kg) for 7 consecutive days, followed by ulcer induction using absolute ethanol (0.2 mL/20 g body weight). Treatment with Co had a remarkable gastroprotection compared to the ethanol-ulcerated mice that significantly reduced the ulcerative lesion index (ULI) and histopathological damage. Daily intragastric administration of Co exerted a powerful anti-inflammatory activity as evidenced by the suppression of nuclear factor (NF)-κB, tumor necrosis factor (TNF)-α, nitric oxide (NO), inducible nitric oxide synthase (iNOS), cyclooxygenase (COX)-2, as well as increased interleukin (IL)-10. Also, pretreatment with Co effectively inhibited ethanol-induced malondialdehyde (MDA) overproduction, increased the depleted superoxide dismutase (SOD) and promoted gastric mucosa epithelial cell proliferation by up-regulating proliferating cell nuclear antigen (PCNA) expression. Similarly, De had a protective effect on ethanol-induced ulcer, which was dependent on the inhibition of inflammatory cytokines and MDA generation, but independent of IL-10, SOD and PCNA improvement. Conclusively, the results have clearly demonstrated the anti-ulcerogenic potential of Co and De on ethanol-induced gastric ulcer; nevertheless, the gastroprotective activity of Co was superior to De due to more multi-pathway regulation than De. These findings suggested that Co or De could be a new useful natural gastroprotective tool against gastric ulcer, which provided a scientific basis for the gastroprotection of sesquiterpene lactones.
Collapse
Affiliation(s)
- Hong Zheng
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Yuling Chen
- Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Jingze Zhang
- Special Drugs R & D Center of People's Armed Police Forces, Department of Pharmacy, Logistics University of Chinese People's Armed Police Forces, Tianjin 300162, China
| | - Lei Wang
- Tianjin Lerentang Pharmaceutical Factory, Tianjin Zhongxin Pharmaceutical Group Co., Ltd., Tianjin 300380, China
| | - Zhaoxiang Jin
- Tianjin Lerentang Pharmaceutical Factory, Tianjin Zhongxin Pharmaceutical Group Co., Ltd., Tianjin 300380, China
| | - Hanhan Huang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Shuli Man
- College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Wenyuan Gao
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China.
| |
Collapse
|
25
|
Carvalho NS, Silva MM, Silva RO, Nicolau LAD, Araújo TSL, Costa DS, Sousa NA, Souza LKM, Soares PMG, Medeiros JVR. Protective Effects of Simvastatin Against Alendronate-Induced Gastric Mucosal Injury in Rats. Dig Dis Sci 2016; 61:400-9. [PMID: 26403426 DOI: 10.1007/s10620-015-3890-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 09/17/2015] [Indexed: 01/05/2023]
Abstract
BACKGROUND It has been reported that simvastatin, a statin commonly prescribed for its anti-inflammatory and antioxidant effects, has gastroprotective effects in indomethacin and ethanol-induced gastric ulcers. However, the effects of simvastatin on alendronate-induced gastric mucosal injury remain unexplored. AIM This study investigated the use of simvastatin for the treatment of alendronate-induced gastric ulcers in rats. METHODS Female rats were pretreated with vehicle or simvastatin (20 and 60 mg/kg p.o.). After 1 h, the rats received alendronate (50 mg/kg p.o.). Simvastatin was administered once daily for 7 days, and from the fourth day of simvastatin treatment, alendronate was administered once daily for 4 days. On the final day of treatment, 4 h after alendronate administration, animals were euthanized, their stomachs were removed, and gastric damage was measured. Samples of the stomach were fixed in 10 % formalin immediately after their removal for subsequent histopathological assessment. Unfixed samples were weighed, frozen at -80 °C until assayed for glutathione (GSH), malondialdehyde (MDA), and cytokine levels and myeloperoxidase (MPO) activity. A third group was used to measure mucus and gastric secretion. RESULTS Pretreatment with simvastatin prevented alendronate-induced macroscopic gastric damage and reduced the levels of MDA and GSH, TNF-α and IL-1β, MPO activity, and mucus levels, in the stomach. CONCLUSIONS This study demonstrates the protective effects of simvastatin against alendronate-induced gastric ulceration. Maintenance of mucosal integrity, inhibition of neutrophil activity, and reduced oxidative stress associated with decreased gastric acidity may explain the gastroprotective effects of simvastatin.
Collapse
Affiliation(s)
- Nathalia S Carvalho
- Post Graduation Program in Pharmacology, Medicinal Plant Research Center (NPPM), Federal University of Piauí, Teresina, PI, Brazil
| | - Mônica M Silva
- Post Graduation Program in Biotechnology, Biotechnology and Biodiversity Center Research (BIOTEC), Federal University of Piauí, Parnaíba, PI, Brazil
| | - Renan O Silva
- Laboratory of Pharmacology of Inflammation and Cancer (LAFICA), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Lucas A D Nicolau
- Laboratory of Pharmacology of Inflammation and Cancer (LAFICA), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Thiago S L Araújo
- Post Graduation Program in Biotechnology, Biotechnology and Biodiversity Center Research (BIOTEC), Federal University of Piauí, Parnaíba, PI, Brazil
| | - Douglas S Costa
- Post Graduation Program in Pharmacology, Medicinal Plant Research Center (NPPM), Federal University of Piauí, Teresina, PI, Brazil
| | - Nayara A Sousa
- Post Graduation Program in Biotechnology, Biotechnology and Biodiversity Center Research (BIOTEC), Federal University of Piauí, Parnaíba, PI, Brazil
| | - Luan K M Souza
- Post Graduation Program in Biotechnology, Biotechnology and Biodiversity Center Research (BIOTEC), Federal University of Piauí, Parnaíba, PI, Brazil
| | - Pedro M G Soares
- Laboratory of Pharmacology of Inflammation and Cancer (LAFICA), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Jand Venes R Medeiros
- Post Graduation Program in Pharmacology, Medicinal Plant Research Center (NPPM), Federal University of Piauí, Teresina, PI, Brazil. .,Post Graduation Program in Biotechnology, Biotechnology and Biodiversity Center Research (BIOTEC), Federal University of Piauí, Parnaíba, PI, Brazil. .,BIOTEC/LAFFEX/UFPI, Av. São Sebastião, no. 2819, Parnaíba, PI, CEP 64202-020, Brazil.
| |
Collapse
|
26
|
Suo H, Feng X, Zhu K, Wang C, Zhao X, Kan J. Shuidouchi (Fermented Soybean) Fermented in Different Vessels Attenuates HCl/Ethanol-Induced Gastric Mucosal Injury. Molecules 2015; 20:19748-63. [PMID: 26540032 PMCID: PMC6332132 DOI: 10.3390/molecules201119654] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 10/20/2015] [Indexed: 12/27/2022] Open
Abstract
Shuidouchi (Natto) is a fermented soy product showing in vivo gastric injury preventive effects. The treatment effects of Shuidouchi fermented in different vessels on HCl/ethanol-induced gastric mucosal injury mice through their antioxidant effect was determined. Shuidouchi contained isoflavones (daidzein and genistein), and GVFS (glass vessel fermented Shuidouchi) had the highest isoflavone levels among Shuidouchi samples fermented in different vessels. After treatment with GVFS, the gastric mucosal injury was reduced as compared to the control mice. The gastric secretion volume (0.47 mL) and pH of gastric juice (3.1) of GVFS treated gastric mucosal injury mice were close to those of ranitidine-treated mice and normal mice. Shuidouchi could decrease serum motilin (MTL), gastrin (Gas) level and increase somatostatin (SS), vasoactive intestinal peptide (VIP) level, and GVFS showed the strongest effects. GVFS showed lower IL-6, IL-12, TNF-α and IFN-γ cytokine levels than other vessel fermented Shuidouchi samples, and these levels were higher than those of ranitidine-treated mice and normal mice. GVFS also had higher superoxide dismutase (SOD), nitric oxide (NO) and malonaldehyde (MDA) contents in gastric tissues than other Shuidouchi samples. Shuidouchi could raise IκB-α, EGF, EGFR, nNOS, eNOS, Mn-SOD, Gu/Zn-SOD, CAT mRNA expressions and reduce NF-κB, COX-2, iNOS expressions as compared to the control mice. GVFS showed the best treatment effects for gastric mucosal injuries, suggesting that glass vessels could be used for Shuidouchi fermentation in functional food manufacturing.
Collapse
Affiliation(s)
- Huayi Suo
- College of Food Science, Southwest University, Chongqing 400715, China.
- Chongqing Engineering Research Center of Regional Food, Chongqing 400715, China.
| | - Xia Feng
- Department of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, China.
- Chongqing Collaborative Innovation Center of Functional Food, Chongqing University of Education, Chongqing 400067, China.
| | - Kai Zhu
- Department of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, China.
- Chongqing Collaborative Innovation Center of Functional Food, Chongqing University of Education, Chongqing 400067, China.
| | - Cun Wang
- Department of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, China.
- Chongqing Collaborative Innovation Center of Functional Food, Chongqing University of Education, Chongqing 400067, China.
| | - Xin Zhao
- Department of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, China.
- Chongqing Collaborative Innovation Center of Functional Food, Chongqing University of Education, Chongqing 400067, China.
| | - Jianquan Kan
- College of Food Science, Southwest University, Chongqing 400715, China.
- Chongqing Engineering Research Center of Regional Food, Chongqing 400715, China.
| |
Collapse
|