Skip to main content
Advertisement

Main menu

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET

User menu

  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Journal of Pharmacology and Experimental Therapeutics
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET
  • My alerts
  • Log in
  • My Cart
Journal of Pharmacology and Experimental Therapeutics

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Visit jpet on Facebook
  • Follow jpet on Twitter
  • Follow jpet on LinkedIn
Research ArticleInflammation, Immunopharmacology, and Asthma

NLRP3 Inflammasome Involvement in the Organ Damage and Impaired Spermatogenesis Induced by Testicular Ischemia and Reperfusion in Mice

Letteria Minutoli, Pietro Antonuccio, Natasha Irrera, Mariagrazia Rinaldi, Alessandra Bitto, Herbert Marini, Gabriele Pizzino, Carmelo Romeo, Antonina Pisani, Giuseppe Santoro, Domenico Puzzolo, Carlo Magno, Francesco Squadrito, Antonio Micali and Domenica Altavilla
Journal of Pharmacology and Experimental Therapeutics December 2015, 355 (3) 370-380; DOI: https://doi.org/10.1124/jpet.115.226936
Letteria Minutoli
Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (C.M.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Pietro Antonuccio
Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (C.M.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Natasha Irrera
Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (C.M.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Mariagrazia Rinaldi
Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (C.M.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Alessandra Bitto
Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (C.M.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Herbert Marini
Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (C.M.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Gabriele Pizzino
Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (C.M.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Carmelo Romeo
Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (C.M.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Antonina Pisani
Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (C.M.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Giuseppe Santoro
Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (C.M.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Domenico Puzzolo
Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (C.M.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Carlo Magno
Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (C.M.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Francesco Squadrito
Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (C.M.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Antonio Micali
Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (C.M.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Domenica Altavilla
Department of Clinical and Experimental Medicine, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (L.M., N.I., M.R., A.B., H.M., G.P., F.S.); Department of Paediatric, Gynaecological, Microbiological, and Biomedical Sciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (P.A., C.R., D.A.); Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (A.P., G.S., D.P., A.M.); and Department of Human Pathology, University of Messina, Azienda Ospedaliera Universitaria Policlinico “G. Martino”, Messina, Italy (C.M.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF
Loading

Abstract

We investigated the role of the nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain containing 3 (NLRP3) inflammasome during testis ischemia and reperfusion injury (TI/R) in wild-type (WT) and NLRP3 knock-out (KO) mice. WT and KO mice underwent 1 hour testicular ischemia followed by 4 hours and 1 and 7 days of reperfusion or a sham TI/R. Furthermore, two groups of WT mice were treated at the beginning of reperfusion and up to 7 days with two inflammasome inhibitors, BAY 11-7082 (20 mg/kg i.p.) or Brilliant Blue G (45.5 mg/kg i.p.), or vehicle. Animals were killed with a pentobarbital sodium overdose at 4 hours and 1 and 7 days, and bilateral orchidectomies were performed. Biochemical and morphologic studies were carried out in all groups. TI/R in WT mice significantly increased caspase-1 and interleukin (IL)-1β mRNA after 4 hours and IL-18 mRNA at 1 day of reperfusion (P ≤ 0.05). There was also a significant increase in caspase-3 and terminal deoxynucleotidyl transferase–mediated digoxigenin-deoxyuridine nick-end labeling–positive cells, marked histologic damage, and altered spermatogenesis in WT mice in both testes after 1 and 7 days of reperfusion. KO TI/R mice, WT TI/R BAY 11-7082, and Brilliant Blue G treated mice showed a significant reduced IL-1β and IL-18 mRNA expression, blunted caspase-1 and -3 expression, minor histologic damages, low terminal deoxynucleotidyl transferase–mediated digoxigenin-deoxyuridine nick-end labeling activity, and preserved spermatogenesis. These data suggest that the activation of NLRP3 plays a key role in TI/R, and its inhibition might represent a therapeutic target for the management of patients with unilateral testicular torsion.

Introduction

Testis torsion is one of the most common testicular lesions in the pediatric population (Fan et al., 2013), with an estimated incidence of 3.5 per 100,000 person-year (Huang et al., 2013). It causes peculiar structural damages, thus leading to infertility (Huang et al., 2013); therefore, it must be treated promptly to avoid ipsilateral testicular dysfunction (Anderson and Williamson, 1986). Events occurring during spontaneous or experimental testicular torsion and subsequent detorsion basically depend on the extension of ischemia (Filho et al., 2004) and are representative of the ischemia and reperfusion injury observed in other organs (Grace, 1994). Under these circumstances, the testis produces several proinflammatory cytokines, including tumor necrosis factor α and interleukin (IL)-1β (Lysiak et al., 2000). The increased levels of these cytokines can be considered indirect evidence of tissue inflammation.

In addition, testicular ischemia/reperfusion (TI/R) triggers the apoptosis cascade, leading to the degeneration of the germinal epithelium (Tripathi et al., 2009). Apoptosis is required for normal spermatogenesis in mammals at different stages of differentiation to ensure cellular homeostasis (Blanco-Rodríguez, 1998). After TI/R, increased expression of Bax and caspase-1 and -3 were observed (Lysiak, 2004), indicating that both initiator and executioner caspases were involved in the apoptotic cascade triggered by the experimental procedure. It was suggested that the regulation of germ cell apoptosis might be central in the development of experimental strategies to prevent their damage (Minutoli et al., 2009).

Inflammasomes are multimolecular complexes assembled in response to various activators and leading to the intracellular activation of the inflammatory cascade, particularly by raising the cleavage of the inactive precursor of IL-1β to its active form (Schroder and Tschopp, 2010). Several inflammasomes have been identified (Tian et al., 2009; Duéñez-Guzmán and Haig, 2014). The nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain containing 3 (NLRP3) inflammasome is currently the most fully characterized, and it is formed by the NOD-like receptor NLRP3 by an adapter protein called apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) and procaspase-1. Furthermore, NLRP3 is of special interest because it responds to different physical and chemical stimuli, leading to many diseases when out of control (Chen and Sun, 2013), and can be assembled following a wide variety of stimuli, including tissue damage (Gross et al., 2011). The actual activation of the NLRP3 inflammasome is a poorly understood event, which involves the integration of many signals indicative of tissue damage or stress (Gross et al., 2011). The inflammasome NLRP3 is an important regulator of caspase-1 (Martinon et al., 2002; Stutz et al., 2009; Kayagaki et al., 2011), which can trigger the sequential activation of caspase-3 (Stutz et al., 2009) and regulate the maturation of the proinflammatory cytokines IL-1β and IL-18 or the rapid inflammatory form of cell death called pyroptosis.

Specific small molecule inhibitors, such as BAY 11-7082 (Juliana et al., 2010) and Brilliant Blue G (BBG) (Zhao et al., 2013), are able to inhibit the proinflammatory and proapoptotic effects of the NLRP3 inflammasome. More in detail, BAY 11-7082, a kinase-β inhibitor, selectively inhibited NLRP3 inflammasome activity in macrophages independently of its inhibitory effect on the priming step following nuclear factor κB activation (Juliana et al., 2010). BBG blocks the membrane-bound purinergic P2X7 receptor (Díaz-Hernández et al., 2009), which in turn suppresses the expression of ASC and inhibits NLRP3 inflammasome activation (Zhao et al., 2013).

In light of this background and to better understand the role of the NLRP3 inflammasome during testicular torsion, we investigated TI/R injury in wild-type (WT) and NLRP3 knock-out (KO) mice.

Materials and Methods

Experimental Protocol.

All animals were housed and maintained under specific pathogen-free conditions at the animal facility of the School of Medicine at the University of Messina, Italy. All experimental procedures complied with the Declaration of Helsinki, with the Guide for the Care and Use of Laboratory Animals, as adopted and promulgated by the United States National Institutes of Health and the Animal Research: Reporting of In Vivo Experiments guidelines (Kilkenny et al., 2010) and were authorized by our local institution. WT (C57Bl6) male mice and KO (Nlrp3tm1bhk) (25–30 g) mice were purchased from the Jackson Laboratory (Bar Harbor, ME). The animals were provided a standard diet ad libitum, with free access to tap water, and were maintained on a 12-hour light/dark cycle. Both WT and KO mice (total number = 126; each group consisted of seven animals) were anesthetized with an intraperitoneal injection of 80 mg/kg of pentobarbital sodium and torsion of the left testis and spermatic cord was performed as previously described (Minutoli et al., 2009). Then, the same testis was detorted. WT and KO animals of all experimental groups underwent testicular ischemia for 1 hour, followed by 4 hours and 1 day and 7 days reperfusion (TI/R). WT and KO sham mice were operated as previously indicated for experimental groups, except for testicular torsion and consequent ischemia. Furthermore, only WT mice were treated either with BAY 11-7082 (20 mg/kg i.p.) (Bitto et al., 2014) or BBG (45.5 mg/kg i.p.) every 2 days (Díaz-Hernández et al., 2009) or vehicle (a 1:3 solution of dimethylsulfoxide and 0.9% NaCl) for up to 7 days. Animals were killed at the above indicated time points after reperfusion with an overdose of pentobarbital sodium, and bilateral orchidectomies were performed. The testes were collected, weighted, and treated for the different experimental technical procedures.

Drugs and Chemicals.

The following compounds were supplied as indicated: BAY 11-7082 by Adipogene (San Diego, CA), BBG by Sigma-Aldrich (Milan, Italy), and sodium pentobarbital by Intervet (Milan, Italy).

Isolation of Soluble Proteins.

Isolation of soluble proteins from testis samples (about 30 mg) of mice was performed in 1 ml of lysis buffer [25 mM Tris/HCl (pH 7.4), 1.0 mM EGTA, 1.0 mM EDTA, 0.5 mM phenyl methylsulfonyl fluoride, 1% NP40, and aprotinin, leupeptin, and pepstatin A (10 mg/ml each)] and homogenized with a Dounce homogenizer (Sigma Aldrich, Milan, Italy; Paterniti et al., 2013). The homogenate was centrifuged at 15,000g for 15 minutes, and the supernatant was collected and used for protein determination with the Bio-Rad protein assay kit (Richmond, CA).

Determination of Active Caspase-1 and -3 by Western Blot Analysis.

Total proteins (30 µg) were denatured with reducing buffer [62 mM Tris (pH 6.8), 10% glycerol, 2% sodium dodecyl sulfate, 5% β-mercaptoethanol, and 0.003% bromophenol blue], separated by electrophoresis, and then transferred onto a polyvinylidene difluoride membrane using a transfer buffer [39 mM glycine and 48 mM Tris (pH 8.3)] at 200 mA for 1 hour. To block membrane proteins, 5% nonfat dry milk in Tris-buffered saline (TBS)–0.1% Tween for 1 hour at room temperature was used, followed by three washes with TBS–0.1% Tween for 10 minutes each, and then incubated with a primary antibody for caspase-1 and -3 (Abcam, Cambridge, UK) and diluted 1:1000 in TBS–0.1% Tween overnight at 4°C. The membranes were washed three times for 10 minutes each with TBS–0.1% Tween to eliminate nonspecific bindings and incubated with a specific peroxidase-conjugated secondary antibody (Pierce, Cambridge, UK), diluted 1:2000, for 1 hour at room temperature. After three washes, the membranes were analyzed by the enhanced chemiluminescence system according to the manufacturer’s protocol (Amersham, Buckinghamshire, UK) as previously described (Minutoli et al., 2005). Equal loading of proteins was assessed on stripped blots by immunodetection of β-actin with a rabbit monoclonal antibody (Cell Signaling, Beverly, MA) diluted 1:500 and peroxidase-conjugated goat antirabbit IgG (Pierce) diluted 1:20,000.

The results from each experimental group [sham WT and KO mice, TI/R + vehicle and contralateral (CL) + vehicle WT and KO mice, and BAY 11-7082– and BBG-treated TI/R and CL WT mice] were obtained from seven samples of each experimental group. For each antibody, three different electrophoretic runs were performed at different time points (4 hours for caspase-1 and 1 and 7 days for caspase-3). The results were normalized versus β-actin and expressed as relative integrated intensity compared with those of sham mice measured with the same batch.

Real-Time PCR.

For the gene expression study, the extraction of total mRNA was performed from testis tissue using TRIZOL (Invitrogen, Milan, Italy) under sterile conditions following the manufacturer’s protocol. For each sample, 5 mg of mRNA were reverse transcripted into cDNA and 3 ml of cDNA were amplified in duplicate with the TaqMan Universal PCR Master Mix containing primer and specific probes (TaqMan), and then using the instrument SDS 7300 Real Time PCR (Applied Biosystems, Foster City, CA). The reversed transcribed product was used to quantify the amount of IL-1β and IL-18. The result was expressed as the number of copies of the target gene compared with the housekeeping gene (β-actin).

Histologic Evaluation.

The testes were fixed in 4% paraformaldehyde in 0.2 M phosphate-buffered saline (PBS), dehydrated in graded ethanol, cleared in xylene, and embedded in paraffin (Paraplast; SPI Supplies, West Chester, PA). Five-micrometer sections were stained with hematoxylin and eosin and photographed with a Zeiss Primo Star (Carl Zeiss Inc., Oberkochen, Germany) light microscope. The images were taken with a Canon A620 Powershot camera (Canon Inc., Tokyo, Japan), saved as tagged image format files, printed at the same final magnification, and blindly assessed by two trained observers without knowledge of the previous treatment. The mean and S.D. of the results were recorded. Five microscopic fields, all including two entire seminiferous tubules from 10 not serial sections of each group, were considered. Morphologic evaluation was performed on both the tubular and extratubular compartments.

As to the tubular compartment, the mean seminiferous tubule diameter was calculated by measuring the diameters of 100 separate seminiferous tubules, all showing a circular profile. A Peak Scale Loupe 7x (GWJ Company, Hacienda Heights, CA) micrometer was used as a scale calibration standard to calculate the diameters, which are expressed in micrometers. Furthermore, spermatogenesis was quantified using the Johnsen’s scoring system (Johnsen, 1970), as modified by Erdemir et al. (2012). Briefly, a score of 10 to 1 was given to each tubule according to epithelial maturation: 10, complete spermatogenesis and perfect tubules; 9, many spermatozoa present and disorganized spermatogenesis; 8, only a few spermatozoa present; 7, no spermatozoa, but many spermatids present; 6, only a few spermatids present; 5, no spermatozoa or spermatids, but many spermatocytes present; 4, only a few spermatocytes present; 3, only spermatogonia present; 2, no germ cells, but only Sertoli cells present; and 1, no germ cells and no Sertoli cells present.

As to the extratubular compartment, a previously described method (Minutoli et al., 2005) for the evaluation of edema, hemorrhagic extravasation, vessel dilation, and Leydig cell change evaluation was used, and each parameter was quantified on the basis of the following scale: 0, absent; 1, mild; 2, moderate; and 3, severe.

Terminal Deoxynucleotidyl Transferase–Mediated Digoxigenin-Deoxyuridine Nick-End Labeling Technique.

For the terminal deoxynucleotidyl transferase–mediated digoxigenin-deoxyuridine nick-end labeling (TUNEL) technique, an apoptosis detection kit (TUNEL Universal Apoptosis Detection Kit; GenScript, Piscataway, NJ) was used on paraffin-embedded 5-μm sections cleared with xylene and dehydrated in graded ethanol. Protein was digested with proteinase K (20 μg/ml PBS), and endogenous peroxidase activity was blocked with 3% H2O2 in methanol. Sections were incubated with terminal deoxynucleotidyl transferase enzyme and biotin-11-day UTP, then with the streptavidin-peroxidase substrate, and finally with the diaminobenzidine solution. Two trained observers without knowledge of the treatment blindly assessed the micrographs, which were obtained and processed as previously indicated for the histologic specimens. From each group, the percentage of tubules with apoptotic cells and the apoptotic index (mean number of TUNEL-positive cells per tubule in 100 seminiferous tubules) (Tsounapi et al., 2012) were calculated.

Statistical Analyses.

All data are expressed as the mean ± S.E., with the exception of the percentage of tubules with apoptotic cells, where absolute values are provided. Data were analyzed with one-way analysis of variance using Tukey’s test as the post hoc procedure or Student’s t test. A P value of ≤0.05 was considered statistically significant.

Results

Active Caspase-1 and Caspase-3 Expression in TI/R.

No caspase-1 activity was demonstrated in the testes of either sham WT or KO mice (Fig. 1A). After 4 hours of reperfusion, TI/R injury determined a significant increase of caspase-1 expression in WT animals. This enhanced activity was higher in the ipsilateral testes and lower in the CL testes (Fig. 1A). Caspase-1 expression was significantly lower in the ipsilateral and CL testes of WT mice treated with both BAY 11-7082 and BBG and in KO mice (Fig. 1A).

Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

Representative Western blot analysis of caspase-1 at 4 hours (A) and caspase-3 at 1 (B) and 7 days (C) in testes of sham WT and KO mice, TI/R + vehicle and CL + vehicle WT and KO mice, and BAY 11-7082– and BBG-treated TI/R and CL WT mice. *P < 0.05 versus sham WT mice; **P < 0.05 versus WT I/R + vehicle mice; #P < 0.05 versus sham KO mice. Bars represent the mean ± S.E. of seven experiments.

No caspase-3 expression was observed in the testes of both sham WT and KO mice at 1 and 7 days (Fig. 1, B and C). On the contrary, TI/R injury induced an increased expression of caspase-3 in the ipsilateral testis of WT animals after 1 and 7 days of reperfusion (Fig. 1, B and C). At 7 days, the levels of caspase-3 further increased when compared with those at 1 day (Fig. 1, B and C). Caspase-3 expression was significantly lower in the ipsilateral testes of WT mice treated with both BAY 11-7082 and BBG and in KO mice (Fig. 1, B and C). Similar results were observed in the CL testes of WT and KO mice even if the absolute values were lower.

IL-1β mRNA and IL-18 mRNA Expression in TI/R.

Testes from sham animals of both strains had negligible basal levels of IL-1β mRNA expression (Fig. 2A). TI/R injury induced an increased expression of IL-1β mRNA in the ipsilateral testes of WT animals following 4 hours of reperfusion (Fig. 2A). IL-1β mRNA expression was significantly lower in the ipsilateral testes of WT mice treated with both BAY 11-7082 and BBG and in KO mice (Fig. 2A). Similar results were observed in the CL testes of WT and KO mice even if the absolute values were lower.

Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

mRNA expression for IL-1β at 4 hours (A) and IL-18 at 1 day (B) in the testes of sham WT and KO mice, TI/R + vehicle and CL + vehicle WT and KO mice, and BAY 11-7082– and BBG-treated TI/R and CL WT mice. *P < 0.05 versus sham WT mice; **P < 0.05 versus WT I/R + vehicle mice; #P < 0.05 versus sham KO mice. Bars represent the mean ± S.E. of seven experiments.

Testes from sham animals of both strains had very low basal levels of IL-18 mRNA expression (Fig. 2B). After 1 day of reperfusion, the expression of IL-18 mRNA was significantly increased in WT animals (Fig. 2B). IL-18 mRNA expression was significantly lower in the ipsilateral testes of WT mice treated with both BAY 11-7082 and BBG and in KO mice (Fig. 2B). Similar results were observed in the CL testes of WT and KO mice even if the absolute values were lower.

Morphologic Evaluation of Apoptosis.

No TUNEL-positive germ cells were observed in the seminiferous tubules from the sham groups of both the WT and KO mice at 1 and 7 days of reperfusion (Fig. 3, A and F; Tables 1A and 2A).

Fig. 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 3.

Assessment of apoptosis with TUNEL technique at 1 and 7 days after the surgical procedure in TI/R WT and KO mice. (A) At 1 day, both sham WT and KO mice testes show no TUNEL-positive cells. (B) In TI/R + vehicle WT mice at 1 day, many TUNEL-positive germ cells (arrows) are present in the seminiferous tubules. (C) In TI/R KO mice at 1 day, some TUNEL-positive germ cells (arrow) are gathered in small groups. (D) In BAY 11-7082–treated TI/R WT mice at 1 day, few TUNEL-positive cells (arrow) are placed in the periphery of the tubules. (E) In BBG-treated TI/R WT mice at 1 day, isolated TUNEL-positive cells (arrow) are evident. (F) At 7 days, in sham WT and KO mice testes, no TUNEL-positive cells are observed. (G) In TI/R + vehicle WT mice at 7 days, some peripheral groups of TUNEL-positive germ cells are evident (arrows). (H) In TI/R KO mice at 7 days, occasional TUNEL-positive germ cells (arrows) are present in the outer part of the tubules. (I) In TI/R BAY 11-7082–treated WT mice at 7 days, rare TUNEL-positive germ cells (arrow) are observed. (J) In TI/R BBG-treated WT mice at 7 days, few groups of TUNEL-positive germ cells (arrow) are present (scale bar: 50 µm).

View this table:
  • View inline
  • View popup
TABLE 1A

Effects of ischemia/reperfusion on the tubular compartment in the testes of WT and KO sham, TI/R and CL WT plus vehicle, BAY 11-7082– and BBG-treated WT, and KO mice at 1 day after the surgical procedure

Values are expressed as the mean ± S.E., with the exception of %TWAC, for which absolute values are provided.

View this table:
  • View inline
  • View popup
TABLE 2A

Effects of ischemia/reperfusion on the tubular compartment in the testes of WT and KO sham, TI/R and CL of WT plus vehicle, BAY 11-7082– and BBG-treated WT, and KO mice at 7 days after the surgical procedure

Values are expressed as the mean ± S.E., with the exception of %TWAC, for which absolute values are provided.

After 1 day of reperfusion, in TI/R WT mice, many TUNEL-positive germ cells were observed in the wall of the seminiferous tubules (Fig. 3B; Table 1A). In TI/R KO mice, few TUNEL-positive germ cells were present in the periphery of the seminiferous tubules (Fig. 3C; Table 1A). Similarly, in BAY 11-7082– (Fig. 3D; Table 1A) and BBG-treated TI/R WT mice (Fig. 3D; Table 1A), testes showed a low number of TUNEL-positive cells only in the peripheral layer of the tubules.

After 7 days of reperfusion, in TI/R WT mice, many peripheral TUNEL-positive germ cells were evident (Fig. 3G; Table 2A). On the contrary, in TI/R KO mice (Fig. 3H; Table 2A) and BAY 11-7082– (Fig. 3I; Table 2A) and BBG-treated TI/R WT mice (Fig. 3J; Table 2A), few isolated TUNEL-positive germ cells were observed in the periphery of the seminiferous tubules.

No significant variations were observed in the CL testes of both strains of mice (morphologic data not shown).

Histologic Evaluation.

After 1 day of reperfusion, the sham groups of both the WT and KO mice showed seminiferous tubules and extratubular compartments with normal morphology (Fig. 4, A and B; Tables 1A and 1B).

Fig. 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 4.

Histologic findings at 1 day after the surgical procedure in seminiferous tubules stained with hematoxylin and eosin. (A and B) Sham WT and KO mice show a normal morphology. (C) In TI/R + vehicle WT mice, degenerative changes of the germinal cells (arrowhead) and edema of the extratubular compartment (*) are evident. (D) CL testes of WT mice show disorganized germinal epithelium (arrows) and hemorrhagic extravasation (h). (E, G, and I) BAY 11-7082– and BBG-treated WT TI/R and KO TI/R testes. Only mild changes of the germinal epithelium and a normal extratubular compartment are present. (F, H, and J) BAY 11-7082– and BBG-treated WT CL and KO CL testes. No evident morphologic changes are recognized in both the tubular and extratubular compartments (scale bar: 50 µm).

View this table:
  • View inline
  • View popup
TABLE 1B

Effects of ischemia/reperfusion on the extratubular compartment in the testes of WT and KO sham, TI/R and CL of WT plus vehicle, BAY 11-7082– and BBG-treated WT, and KO mice at 1 day after the surgical procedure

Values are expressed as the mean ± S.E. Histologic grading of the extratubular compartment changes was based on the following scale: 0, absent; 1, mild; 2, moderate; and 3, severe.

In the WT mice, the TI/R group showed massive tubular disorganization, with degenerative changes of the germinal cells, interstitial edema, hemorrhagic extravasation from slightly dilated vessels, and mild Leydig cell changes (Fig. 4C; Tables 1A and 1B). CL testes of the same mice showed small tubules with disorganized germinal epithelium and vascular dilation with hemorrhagic extravasation (Fig. 4D; Tables 1A and 1B).

In BAY 11-7082– (Fig. 4E; Tables 1A and 1B) and BBG-treated WT mice (Fig. 4G; Tables 1A and 1B) and KO mice (Fig. 4I; Tables 1A and 1B), TI/R testes showed only mild changes of the germinal epithelium, but a normal extratubular compartment. In the same groups (Fig. 4, F, H, and J; Tables 1A and 1B), CL testes showed well preserved sperm cells and a normal extratubular compartment.

After 7 days of reperfusion, the sham groups showed normal morphology of both the seminiferous tubules and extratubular compartment (Fig. 5, A and B; Tables 2A and 2B).

Fig. 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 5.

Histologic findings at 7 days after the surgical procedure in seminiferous tubules stained with hematoxylin and eosin. (A and B) Sham WT and KO mice show a normal morphology. (C) TI/R + vehicle WT mice. Tubules with disorganized germinal epithelium (arrow) and a widened extratubular compartment (*) are evident. (D) CL testes of WT mice. A well evident edema (*) is present in the extratubular compartment. (E, G, and I) BAY 11-7082–and BBG-treated WT TI/R and KO TI/R testes. Many immature spermatids (s), some spermatozoa (z), occasional mitoses (m), and intercellular clefts (arrow) are evident. The extratubular compartment exhibits mild edema and few dilated vessels. (F, H, and J) BAY 11-7082– and BBG-treated WT CL and KO CL testes. Better preserved sperm cells and a normal extratubular compartment are present (scale bar: 50 µm).

View this table:
  • View inline
  • View popup
TABLE 2B

Effects of ischemia/reperfusion on the extratubular compartment in the testes of WT and KO sham, TI/R and CL of WT plus vehicle, BAY 11-7082– and BBG-treated WT, and KO mice at 7 days after the surgical procedure

Values are expressed as the mean ± S.E. Histologic grading of the extratubular compartment changes was based on the following scale: 0, absent; 1, mild; 2, moderate; and 3, severe.

In the WT mice, the TI/R group presented small tubules with a highly disorganized germinal epithelium and some spermatogonia along their periphery. The extratubular compartment was damaged, and Leydig cells showed mild-to-moderate structural changes consisting of pale cytoplasm and darker and condensed nuclei (Fig. 5C; Tables 2A and 2B). CL testes of WT mice at the same time had larger, less damaged tubules, while the extratubular compartment showed well evident edema (Fig. 5D; Tables 2A and 2B).

In BAY 11-7082– (Fig. 5E; Tables 2A and 2B) and BBG-treated WT mice (Fig. 5G; Tables 2A and 2B) and KO mice (Fig. 5I; Tables 2A and 2B), TI/R testes showed a germinal epithelium, with many immature spermatids, reduced spermatozoa, and some intercellular clefts. The extratubular compartment exhibited mild edema and few dilated vessels. In the same groups (Fig. 5, F, H, and J; Tables 2A and 2B), CL testes showed better preserved sperm cells and a normal extratubular compartment.

Discussion

Experimental studies have suggested that innate pattern recognition receptors may prime tissue and organ damage during ischemic states (Shigeoka et al., 2007, 2010; Haneklaus et al., 2013).

Testicular torsion is characterized by tissue damage and represents a urological emergency (Ringdahl and Teague, 2006), which, when misdiagnosed and inappropriately treated, can lead to male infertility (Antonuccio et al., 2006). Several mechanisms have been implicated in the development of testicular damage following the torsion and detorsion processes (Lysiak et al., 2000; Nadiri et al., 2006; Srinivasan et al., 2007; Minutoli et al., 2009, 2012), which have been considered similar to ischemia-reperfusion injuries observed in different organs (Eltzschig and Eckle, 2011).

Ischemia and reperfusion is a pathologic event characterized, at first, by a reduction of blood supply followed by the reestablishment of perfusion and related reoxygenation (Eltzschig and Eckle, 2011). It induces early tissue injuries, such as reactive oxygen species (ROS) generation (Lei et al., 2015), interstitial edema (Widgerow, 2014), and a damaged barrier function of endothelial cells, due to reduced adenylate cyclase activity and intracellular cAMP levels. In particular, during ischemia and reperfusion, nucleotides in the form of ATP promote tissue inflammation and activate the NLRP3 inflammasome (McDonald et al., 2010).

Ischemia and reperfusion activates various programs of cell death, among which necrosis and apoptosis play a significant role. Necrosis is characterized by a swelling of cells and organelles, with rupture of their membranes and release of their contents. These cells trigger the inflammatory process and cytokine production. In contrast, apoptosis is characterized by a cellular and nuclear shrinkage and plasma membrane integrity (Hotchkiss et al., 2009).

In particular, TI/R results in exaggerated production of ROS, activates the mitogen-activated protein kinases family, and triggers the inflammatory cascade and apoptosis machinery. This pathologic cascade is responsible for the testicular atrophy and impaired spermatogenesis observed at a later stage (Filho et al., 2004; Antonuccio et al., 2006; Nadiri et al., 2006; Srinivasan et al., 2007; Minutoli et al., 2009, 2012). Even if the importance of early events is well recognized, the late phase of TI/R is crucial from a clinical point of view as the testis morphology and function could be severely impaired (Altavilla et al., 2012; Cvetkovic et al., 2015).

Increased levels of tumor necrosis factor α and IL-1β have been measured during testicular ischemia and reperfusion (Lysiak et al., 2000), thus confirming a role of inflammation in testis twisting. Inflammation works in concert with apoptosis to induce late organ damage and may be responsible for infertility.

Apoptosis, further than a physiologic process that entails the programmed cell death, can also induce testicular injury. In fact, this process is essential during normal spermatogenesis in both humans and animals; however, TI/R injury causes apoptosis of germ cells (Tripathi et al., 2009). All these findings, taken together, clearly suggest that inflammation and apoptosis are important components of the organ damage and impaired spermatogenesis induced by testicular twisting and untwisting.

Muckle-Wells syndrome (MWS) is an inflammatory disease consisting of recurrent symptoms of ocular and joint inflammation, fever, and skin rash, which is due to NLRP3 gene mutations causing an impairment in the activity of the gene product cryopyrin (Agostini et al., 2004). Cryopyrin induces caspase-1 and, in turn, the cleavage of IL-1β and IL-18 that causes the typical inflammatory disease of MWS (Agostini et al., 2004; Hoffman et al., 2001).

Recently, a study suggested that the NLRP3/ASC/caspase-1 axis regulated the proinflammatory cytokines IL-1β and IL-18 (Xie et al., 2014).

Interestingly, patients suffering from MWS have oligozoospermia and/or azoospermia and show subfertility or infertility (Tran et al., 2012). On the basis of this observation and of other experimental data (Lech et al., 2010), as NLRP3 is involved in the molecular cascade of inflammation and apoptosis in response to cellular ischemia, we thought it was worthy of interest to investigate its role in the altered spermatogenesis induced by TI/R injury.

IL-1β and IL-18 were markedly reduced in the TI/R inflammasome–deficient mice compared with the TI/R WT animals. These data suggest that the inflammatory cascade induced by TI/R injury is blunted in the absence of this molecular platform.

Also, contralateral testes showed evident structural damages, even if they were milder than those of the I/R testes. Several mechanisms have been proposed to understand the contralateral testicular damage, such as a decrease in testicular blood flow after an afferent stimulus (Prillaman and Turner, 1997; Andiran et al., 2000; Minutoli et al., 2005), ROS generation after detorsion and overproduction of nitric oxide (NO), or autoimmunization against the spermatogonia (Harrison et al., 1981; Sarica et al., 1997; Shiraishi et al., 2001). In this context, the role of NO is of primary importance as the endothelial nitric oxide synthase and neuronal nitric oxide synthase are: 1) constitutively expressed in the testis, 2) activated transiently in response to the increase in intracellular Ca2+ mobilization, and 3) contribute to the physiologic regulation of vascular tone in both the testes and other target organs (Shiraishi et al., 2001; Esposito and Cuzzocrea, 2009). Moreover, the delicate balance between NO and peroxynitrite catalysts appears as an interesting therapeutic target in ischemia/reperfusion injury (Esposito and Cuzzocrea, 2009).

In the present study, we also studied caspase-1 and -3 expression after reperfusion in both WT and KO animals. Our results demonstrated that KO mice exhibited a reduced expression of both caspases when compared with WT animals during TI/R. Accordingly, a large number of TUNEL-positive germ cells were observed in the seminiferous tubules from TI/R WT mice either at 1 or 7 days, mostly in the outer layer of the seminiferous tubules, suggesting that apoptosis is acutely increased in the tubular tissue of the twisted testes. NLRP3 KO mice showed a reduced number of TUNEL-positive cells, thus suggesting that the absence of this inflammasome reduces the activation of the apoptotic machinery. Therefore, an important finding of our paper is that NLRP3-deficient mice respond to the ischemia and reperfusion insult, with a lower activation of the inflammatory and apoptosis cascade than in WT animals.

The most susceptible function to testicular twisting is spermatogenesis. In fact, WT animals subjected to TI/R had a very poor Johnsen’s score, which is a method to evaluate spermatogenesis (Johnsen, 1970; Erdemir et al., 2012). Interestingly, the lack of NLRP3 produced an improvement in spermatogenesis following the surgical insult of TI/R. This finding is of particular interest. In fact, it is possible to speculate that this molecular platform represents one of the most prominent mechanisms for the development of the late complications associated with testicular torsion.

To confirm the crucial role of the NLRP3 inflammasome in TI/R, we used another way of activation blocking by means of specific inhibitors, such as BAY 11-7082 (Juliana et al., 2010) and BBG (Zhao et al., 2013), which were administered in TI/R WT mice. Our data showed biochemical and morphologic patterns similar to those of KO mice, thus indicating that the absence of changes can be related to the blockade of either the priming step (Juliana et al., 2010) or the membrane-bound purinergic P2X7 receptor (Díaz-Hernández et al., 2009). Furthermore, our results indicate that NLRP3 is an interesting target for innovative drugs aimed to treat altered spermatogenesis and male fertility and subfertility.

In conclusion, our study is the first to our knowledge to suggest that the NLRP3 inflammasome signals inflammatory and apoptotic responses in the testis during low-flow states.

Acknowledgments

The authors thank Mr. Sebastiano Brunetto from the Department of Biomedical Sciences and Morphofunctional Images of the University of Messina for technical assistance.

Authorship Contributions

Participated in research design: Squadrito, Minutoli, Altavilla, Micali.

Conducted experiments: Puzzolo, Irrera, Rinaldi, Bitto, Santoro, Pisani, Pizzino, Antonuccio.

Performed data analysis: Marini, Magno, Bitto.

Wrote or contributed to the writing of the manuscript: Squadrito, Altavilla, Minutoli, Romeo, Marini, Micali.

Footnotes

    • Received June 17, 2015.
    • Accepted September 24, 2015.
  • This work was supported by departmental funding.

  • dx.doi.org/10.1124/jpet.115.226936.

Abbreviations

ASC
apoptosis-associated speck-like protein containing a caspase recruitment domain
BBG
Brilliant Blue G
CL
contralateral
IL
interleukin
KO
knock out
MWS
Muckle-Wells syndrome
NLRP3
NOD-like receptor family pyrin domain containing 3
NO
nitric oxide
NOD
nucleotide-binding oligomerization domain
PBS
phosphate-buffered saline
ROS
reactive oxygen species
TBS
Tris-buffered saline
TI/R
testicular ischemia/reperfusion
TUNEL
terminal deoxynucleotidyl transferase–mediated digoxigenin-deoxyuridine nick-end labeling
WT
wild type
  • Copyright © 2015 by The American Society for Pharmacology and Experimental Therapeutics

References

  1. ↵
    1. Agostini L,
    2. Martinon F,
    3. Burns K,
    4. McDermott MF,
    5. Hawkins PN, and
    6. Tschopp J
    (2004) NALP3 forms an IL-1beta-processing inflammasome with increased activity in Muckle-Wells autoinflammatory disorder. Immunity 20:319–325.
    OpenUrlCrossRefPubMed
  2. ↵
    1. Altavilla D,
    2. Romeo C,
    3. Squadrito F,
    4. Marini H,
    5. Morgia G,
    6. Antonuccio P, and
    7. Minutoli L
    (2012) Molecular pathways involved in the early and late damage induced by testis ischemia: evidence for a rational pharmacological modulation. Curr Med Chem 19:1219–1224.
    OpenUrlCrossRefPubMed
  3. ↵
    1. Anderson JB and
    2. Williamson RC
    (1986) The fate of the human testes following unilateral torsion of the spermatic cord. Br J Urol 58:698–704.
    OpenUrlCrossRefPubMed
  4. ↵
    1. Andiran F,
    2. Okur DH,
    3. Kilinç A,
    4. Gedikoḡlu G,
    5. Kilinç K, and
    6. Tanyel FC
    (2000) Do experimentally induced ipsilateral testicular torsion, vas deferens obstruction, intra-abdominal testis or venous obstruction damage the contralateral testis trough a common mechanism? BJU Int 85:330–335.
    OpenUrlCrossRefPubMed
  5. ↵
    1. Antonuccio P,
    2. Minutoli L,
    3. Romeo C,
    4. Nicòtina PA,
    5. Bitto A,
    6. Arena S,
    7. Altavilla D,
    8. Zuccarello B,
    9. Polito F, and
    10. Squadrito F
    (2006) Lipid peroxidation activates mitogen-activated protein kinases in testicular ischemia-reperfusion injury. J Urol 176:1666–1672.
    OpenUrlCrossRefPubMed
  6. ↵
    1. Bitto A,
    2. Altavilla D,
    3. Pizzino G,
    4. Irrera N,
    5. Pallio G,
    6. Colonna MR, and
    7. Squadrito F
    (2014) Inhibition of inflammasome activation improves the impaired pattern of healing in genetically diabetic mice. Br J Pharmacol 171:2300–2307.
    OpenUrlCrossRefPubMed
  7. ↵
    1. Blanco-Rodríguez J
    (1998) A matter of death and life: the significance of germ cell death during spermatogenesis. Int J Androl 21:236–248.
    OpenUrlCrossRefPubMed
  8. ↵
    1. Chen S and
    2. Sun B
    (2013) Negative regulation of NLRP3 inflammasome signaling. Protein Cell 4:251–258.
    OpenUrlCrossRefPubMed
  9. ↵
    1. Cvetkovic T,
    2. Stankovic J,
    3. Najman S,
    4. Pavlovic D,
    5. Stokanovic D,
    6. Vlajkovic S,
    7. Dakovic-Bjelakovic M,
    8. Cukuranovic J,
    9. Zivkovic V, and
    10. Stefanovic V
    (2015) Oxidant and antioxidant status in experimental rat testis after testicular torsion/detorsion. Int J Fertil Steril 9:121–128.
    OpenUrlPubMed
  10. ↵
    1. Díaz-Hernández M,
    2. Díez-Zaera M,
    3. Sánchez-Nogueiro J,
    4. Gómez-Villafuertes R,
    5. Canals JM,
    6. Alberch J,
    7. Miras-Portugal MT, and
    8. Lucas JJ
    (2009) Altered P2X7-receptor level and function in mouse models of Huntington’s disease and therapeutic efficacy of antagonist administration. FASEB J 23:1893–1906.
    OpenUrlAbstract/FREE Full Text
  11. ↵
    1. Duéñez-Guzmán EA and
    2. Haig D
    (2014) The evolution of reproduction-related NLRP genes. J Mol Evol 78:194–201.
    OpenUrlCrossRefPubMed
  12. ↵
    1. Eltzschig HK and
    2. Eckle T
    (2011) Ischemia and reperfusion--from mechanism to translation. Nat Med 17:1391–1401.
    OpenUrlCrossRefPubMed
  13. ↵
    1. Erdemir F,
    2. Atilgan D,
    3. Markoc F,
    4. Boztepe O,
    5. Suha-Parlaktas B, and
    6. Sahin S
    (2012) [The effect of diet induced obesity on testicular tissue and serum oxidative stress parameters]. Actas Urol Esp 36:153–159.
    OpenUrlPubMed
  14. ↵
    1. Esposito E and
    2. Cuzzocrea S
    (2009) Role of nitroso radicals as drug targets in circulatory shock. Br J Pharmacol 157:494–508.
    OpenUrlCrossRefPubMed
  15. ↵
    1. Fan R,
    2. Zhang J,
    3. Cheng L, and
    4. Lin J
    (2013) Testicular and paratesticular pathology in the pediatric population: a 20 year experience at Riley hospital for children. Pathol Res Pract 209:404–408.
    OpenUrlCrossRefPubMed
  16. ↵
    1. Filho DW,
    2. Torres MA,
    3. Bordin AL,
    4. Crezcynski-Pasa TB, and
    5. Boveris A
    (2004) Spermatic cord torsion, reactive oxygen and nitrogen species and ischemia-reperfusion injury. Mol Aspects Med 25:199–210.
    OpenUrlCrossRefPubMed
  17. ↵
    1. Grace PA
    (1994) Ischaemia-reperfusion injury. Br J Surg 81:637–647.
    OpenUrlCrossRefPubMed
  18. ↵
    1. Gross O,
    2. Thomas CJ,
    3. Guarda G, and
    4. Tschopp J
    (2011) The inflammasome: an integrated view. Immunol Rev 243:136–151.
    OpenUrlCrossRefPubMed
  19. ↵
    1. Haneklaus M,
    2. O’Neill LA, and
    3. Coll RC
    (2013) Modulatory mechanisms controlling the NLRP3 inflammasome in inflammation: recent developments. Curr Opin Immunol 25:40–45.
    OpenUrlCrossRefPubMed
  20. ↵
    1. Harrison RG,
    2. Lewis-Jones DI,
    3. Moreno de Marval MJ, and
    4. Connolly RC
    (1981) Mechanism of damage to the contralateral testis in rats with an ischaemic testis. Lancet 2:723–725.
    OpenUrlPubMed
  21. ↵
    1. Hoffman HM,
    2. Mueller JL,
    3. Broide DH,
    4. Wanderer AA, and
    5. Kolodner RD
    (2001) Mutation of a new gene encoding a putative pyrin-like protein causes familial cold autoinflammatory syndrome and Muckle-Wells syndrome. Nat Genet 29:301–305.
    OpenUrlCrossRefPubMed
  22. ↵
    1. Hotchkiss RS,
    2. Strasser A,
    3. McDunn JE, and
    4. Swanson PE
    (2009) Cell death. N Engl J Med 361:1570–1583.
    OpenUrlCrossRefPubMed
  23. ↵
    1. Huang WY,
    2. Chen YF,
    3. Chang HC,
    4. Yang TK,
    5. Hsieh JT, and
    6. Huang KH
    (2013) The incidence rate and characteristics in patients with testicular torsion: a nationwide, population-based study. Acta Paediatr 102:e363–e367.
    OpenUrlCrossRefPubMed
  24. ↵
    1. Johnsen SG
    (1970) Testicular biopsy score count--a method for registration of spermatogenesis in human testes: normal values and results in 335 hypogonadal males. Hormones 1:2–25.
    OpenUrlCrossRefPubMed
  25. ↵
    1. Juliana C,
    2. Fernandes-Alnemri T,
    3. Wu J,
    4. Datta P,
    5. Solorzano L,
    6. Yu JW,
    7. Meng R,
    8. Quong AA,
    9. Latz E,
    10. Scott CP,
    11. et al.
    (2010) Anti-inflammatory compounds parthenolide and Bay 11-7082 are direct inhibitors of the inflammasome. J Biol Chem 285:9792–9802.
    OpenUrlAbstract/FREE Full Text
  26. ↵
    1. Kayagaki N,
    2. Warming S,
    3. Lamkanfi M,
    4. Vande Walle L,
    5. Louie S,
    6. Dong J,
    7. Newton K,
    8. Qu Y,
    9. Liu J,
    10. Heldens S,
    11. et al.
    (2011) Non-canonical inflammasome activation targets caspase-11. Nature 479:117–121.
    OpenUrlCrossRefPubMed
  27. ↵
    1. Kilkenny C,
    2. Browne WJ,
    3. Cuthill IC,
    4. Emerson M, and
    5. Altman DG
    (2010) Improving bioscience research reporting: the ARRIVE guidelines for reporting animal research. PLoS Biol 8:e1000412.
    OpenUrlCrossRefPubMed
  28. ↵
    1. Lech M,
    2. Avila-Ferrufino A,
    3. Skuginna V,
    4. Susanti HE, and
    5. Anders HJ
    (2010) Quantitative expression of RIG-like helicase, NOD-like receptor and inflammasome-related mRNAs in humans and mice. Int Immunol 22:717–728.
    OpenUrlAbstract/FREE Full Text
  29. ↵
    1. Lei X,
    2. Chao H,
    3. Zhang Z,
    4. Lv J,
    5. Li S,
    6. Wei H,
    7. Xue R,
    8. Li F, and
    9. Li Z
    (2015) Neuroprotective effects of quercetin in a mouse model of brain ischemic/reperfusion injury via anti-apoptotic mechanisms based on the Akt pathway. Mol Med Rep 12:3688–3696.
    OpenUrlPubMed
  30. ↵
    1. Lysiak JJ
    (2004) The role of tumor necrosis factor-alpha and interleukin-1 in the mammalian testis and their involvement in testicular torsion and autoimmune orchitis. Reprod Biol Endocrinol 2:9.
    OpenUrlCrossRefPubMed
  31. ↵
    1. Lysiak JJ,
    2. Turner SD, and
    3. Turner TT
    (2000) Molecular pathway of germ cell apoptosis following ischemia/reperfusion of the rat testis. Biol Reprod 63:1465–1472.
    OpenUrlAbstract/FREE Full Text
  32. ↵
    1. Martinon F,
    2. Burns K, and
    3. Tschopp J
    (2002) The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-β. Mol Cell 10:417–426.
    OpenUrlCrossRefPubMed
  33. ↵
    1. McDonald B,
    2. Pittman K,
    3. Menezes GB,
    4. Hirota SA,
    5. Slaba I,
    6. Waterhouse CC,
    7. Beck PL,
    8. Muruve DA, and
    9. Kubes P
    (2010) Intravascular danger signals guide neutrophils to sites of sterile inflammation. Science 330:362–366.
    OpenUrlAbstract/FREE Full Text
  34. ↵
    1. Minutoli L,
    2. Antonuccio P,
    3. Polito F,
    4. Bitto A,
    5. Squadrito F,
    6. Di Stefano V,
    7. Nicotina PA,
    8. Fazzari C,
    9. Maisano D,
    10. Romeo C,
    11. et al.
    (2009) Mitogen-activated protein kinase 3/mitogen-activated protein kinase 1 activates apoptosis during testicular ischemia-reperfusion injury in a nuclear factor-kappaB-independent manner. Eur J Pharmacol 604:27–35.
    OpenUrlCrossRefPubMed
  35. ↵
    1. Minutoli L,
    2. Antonuccio P,
    3. Romeo C,
    4. Nicòtina PA,
    5. Bitto A,
    6. Arena S,
    7. Polito F,
    8. Altavilla D,
    9. Turiaco N,
    10. Cutrupi A,
    11. et al.
    (2005) Evidence for a role of mitogen-activated protein kinase 3/mitogen-activated protein kinase in the development of testicular ischemia-reperfusion injury. Biol Reprod 73:730–736.
    OpenUrlAbstract/FREE Full Text
  36. ↵
    1. Minutoli L,
    2. Antonuccio P,
    3. Squadrito F,
    4. Bitto A,
    5. Nicotina PA,
    6. Fazzari C,
    7. Polito F,
    8. Marini H,
    9. Bonvissuto G,
    10. Arena S,
    11. et al.
    (2012) Effects of polydeoxyribonucleotide on the histological damage and the altered spermatogenesis induced by testicular ischaemia and reperfusion in rats. Int J Androl 35:133–144.
    OpenUrlCrossRefPubMed
  37. ↵
    1. Nadiri A,
    2. Wolinski MK, and
    3. Saleh M
    (2006) The inflammatory caspases: key players in the host response to pathogenic invasion and sepsis. J Immunol 177:4239–4245.
    OpenUrlAbstract/FREE Full Text
  38. ↵
    1. Paterniti I,
    2. Impellizzeri D,
    3. Di Paola R,
    4. Navarra M,
    5. Cuzzocrea S, and
    6. Esposito E
    (2013) A new co-ultramicronized composite including palmitoylethanolamide and luteolin to prevent neuroinflammation in spinal cord injury. J Neuroinflammation 10:91.
    OpenUrlCrossRefPubMed
  39. ↵
    1. Prillaman HM and
    2. Turner TT
    (1997) Rescue of testicular function after acute experimental torsion. J Urol 157:340–345.
    OpenUrlCrossRefPubMed
  40. ↵
    1. Ringdahl E and
    2. Teague L
    (2006) Testicular torsion. Am Fam Physician 74:1739–1743.
    OpenUrlPubMed
  41. ↵
    1. Sarica K,
    2. Küpeli B,
    3. Budak M,
    4. Koşar A,
    5. Kavukçu M,
    6. Durak I, and
    7. Göğüş O
    (1997) Influence of experimental spermatic cord torsion on the contralateral testis in rats. Evaluation of tissue free oxygen radical scavenger enzyme levels. Urol Int 58:208–212.
    OpenUrlCrossRefPubMed
  42. ↵
    1. Schroder K and
    2. Tschopp J
    (2010) The inflammasomes. Cell 140:821–832.
    OpenUrlCrossRefPubMed
  43. ↵
    1. Shigeoka AA,
    2. Holscher TD,
    3. King AJ,
    4. Hall FW,
    5. Kiosses WB,
    6. Tobias PS,
    7. Mackman N, and
    8. McKay DB
    (2007) TLR2 is constitutively expressed within the kidney and participates in ischemic renal injury through both MyD88-dependent and -independent pathways. J Immunol 178:6252–6258.
    OpenUrlAbstract/FREE Full Text
  44. ↵
    1. Shigeoka AA,
    2. Kambo A,
    3. Mathison JC,
    4. King AJ,
    5. Hall WF,
    6. da Silva Correia J,
    7. Ulevitch RJ, and
    8. McKay DB
    (2010) Nod1 and nod2 are expressed in human and murine renal tubular epithelial cells and participate in renal ischemia reperfusion injury. J Immunol 184:2297–2304.
    OpenUrlAbstract/FREE Full Text
  45. ↵
    1. Shiraishi K,
    2. Naito K, and
    3. Yoshida K
    (2001) Nitric oxide promotes germ cell necrosis in the delayed phase after experimental testicular torsion of rat. Biol Reprod 65:514–521.
    OpenUrlAbstract/FREE Full Text
  46. ↵
    1. Srinivasan AK,
    2. Freyle J,
    3. Gitlin JS, and
    4. Palmer LS
    (2007) Climatic conditions and the risk of testicular torsion in adolescent males. J Urol 178:2585–2588, discussion 2588.
    OpenUrlCrossRefPubMed
  47. ↵
    1. Stutz A,
    2. Golenbock DT, and
    3. Latz E
    (2009) Inflammasomes: too big to miss. J Clin Invest 119:3502–3511.
    OpenUrlCrossRefPubMed
  48. ↵
    1. Tian X,
    2. Pascal G, and
    3. Monget P
    (2009) Evolution and functional divergence of NLRP genes in mammalian reproductive systems. BMC Evol Biol 9:202.
    OpenUrlCrossRefPubMed
  49. ↵
    1. Tran TA,
    2. Koné-Paut I,
    3. Marie I,
    4. Ninet J,
    5. Cuisset L, and
    6. Meinzer U
    (2012) Muckle-Wells syndrome and male hypofertility: a case series. Semin Arthritis Rheum 42:327–331.
    OpenUrlCrossRefPubMed
  50. ↵
    1. Tripathi R,
    2. Mishra DP, and
    3. Shaha C
    (2009) Male germ cell development: turning on the apoptotic pathways. J Reprod Immunol 83:31–35.
    OpenUrlCrossRefPubMed
  51. ↵
    1. Tsounapi P,
    2. Saito M,
    3. Dimitriadis F,
    4. Kitatani K,
    5. Kinoshita Y,
    6. Shomori K,
    7. Takenaka A, and
    8. Satoh K
    (2012) The role of K ATP channels on ischemia-reperfusion injury in the rat testis. Life Sci 90:649–656.
    OpenUrlCrossRefPubMed
  52. ↵
    1. Widgerow AD
    (2014) Ischemia-reperfusion injury: influencing the microcirculatory and cellular environment. Ann Plast Surg 72:253–260.
    OpenUrlCrossRefPubMed
  53. ↵
    1. Xie Q,
    2. Shen WW,
    3. Zhong J,
    4. Huang C,
    5. Zhang L, and
    6. Li J
    (2014) Lipopolysaccharide/adenosine triphosphate induces IL‑1β and IL-18 secretion through the NLRP3 inflammasome in RAW264.7 murine macrophage cells. Int J Mol Med 34:341–349.
    OpenUrlPubMed
  54. ↵
    1. Zhao J,
    2. Wang H,
    3. Dai C,
    4. Wang H,
    5. Zhang H,
    6. Huang Y,
    7. Wang S,
    8. Gaskin F,
    9. Yang N, and
    10. Fu SM
    (2013) P2X7 blockade attenuates murine lupus nephritis by inhibiting activation of the NLRP3/ASC/caspase 1 pathway. Arthritis Rheum 65:3176–3185.
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Journal of Pharmacology and Experimental Therapeutics: 355 (3)
Journal of Pharmacology and Experimental Therapeutics
Vol. 355, Issue 3
1 Dec 2015
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Editorial Board (PDF)
  • Front Matter (PDF)
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Journal of Pharmacology and Experimental Therapeutics article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
NLRP3 Inflammasome Involvement in the Organ Damage and Impaired Spermatogenesis Induced by Testicular Ischemia and Reperfusion in Mice
(Your Name) has forwarded a page to you from Journal of Pharmacology and Experimental Therapeutics
(Your Name) thought you would be interested in this article in Journal of Pharmacology and Experimental Therapeutics.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Research ArticleInflammation, Immunopharmacology, and Asthma

Role of NLRP3 Inflammasome in Testicular Ischemia

Letteria Minutoli, Pietro Antonuccio, Natasha Irrera, Mariagrazia Rinaldi, Alessandra Bitto, Herbert Marini, Gabriele Pizzino, Carmelo Romeo, Antonina Pisani, Giuseppe Santoro, Domenico Puzzolo, Carlo Magno, Francesco Squadrito, Antonio Micali and Domenica Altavilla
Journal of Pharmacology and Experimental Therapeutics December 1, 2015, 355 (3) 370-380; DOI: https://doi.org/10.1124/jpet.115.226936

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Research ArticleInflammation, Immunopharmacology, and Asthma

Role of NLRP3 Inflammasome in Testicular Ischemia

Letteria Minutoli, Pietro Antonuccio, Natasha Irrera, Mariagrazia Rinaldi, Alessandra Bitto, Herbert Marini, Gabriele Pizzino, Carmelo Romeo, Antonina Pisani, Giuseppe Santoro, Domenico Puzzolo, Carlo Magno, Francesco Squadrito, Antonio Micali and Domenica Altavilla
Journal of Pharmacology and Experimental Therapeutics December 1, 2015, 355 (3) 370-380; DOI: https://doi.org/10.1124/jpet.115.226936
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgments
    • Authorship Contributions
    • Footnotes
    • Abbreviations
    • References
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • ONO-4641 Alleviates Mouse Immune-Mediated Aplastic Anemia
  • PP2A Is a Novel Therapeutic Target in Gout.
  • Genomic Effects of an EP4-Receptor Agonist in Human Airway Epithelia
Show more Inflammation, Immunopharmacology, and Asthma

Similar Articles

  • Home
  • Alerts
Facebook   Twitter   LinkedIn   RSS

Navigate

  • Current Issue
  • Fast Forward by date
  • Fast Forward by section
  • Latest Articles
  • Archive
  • Search for Articles
  • Feedback
  • ASPET

More Information

  • About JPET
  • Editorial Board
  • Instructions to Authors
  • Submit a Manuscript
  • Customized Alerts
  • RSS Feeds
  • Subscriptions
  • Permissions
  • Terms & Conditions of Use

ASPET's Other Journals

  • Drug Metabolism and Disposition
  • Molecular Pharmacology
  • Pharmacological Reviews
  • Pharmacology Research & Perspectives
ISSN 1521-0103 (Online)

Copyright © 2021 by the American Society for Pharmacology and Experimental Therapeutics