Abstract
Minocycline is a widely used tetracycline antibiotic. It has been used for decades in the treatment of various gram-positive and gram-negative infections. More recently, minocycline has been shown to have neuroprotective properties in different animal models of acute neurological injury. As a neuroprotective agent, minocycline has the potential to be superior to most of the previously tried agents. In addition to its high blood-brain barrier penetration, minocycline is also a safe compound commonly used in chronic infections. Its multiple mechanisms of action (anti-inflammatory, antiapoptotic and protease inhibitor) in neuroprotection make it a desirable candidate therapy for acute neurological injury, such as ischemic stroke. Minocycline is ready for clinical trials in acute neurologic injury.
Keywords: Minocycline, Stroke, neuroprotection
Numerous studies have been conducted in the hope of developing an effective neuroprotective agent for acute brain injury, particularly ischemic stroke. To date, no pharmacologic agent has been shown to be efficacious in a context of acute intervention. It is likely that agents like calcium channel blockers and glutamate antagonists did not show benefit in acute brain injury for several reasons and may include: poor blood-brain barrier penetration; dose limiting toxicity of the agent;1–4 and the time-window in which the drug was effective was not long enough to be translated readily into clinical use.3–6 After careful review of the failed studies, it is clear that there is a need to investigate new approaches to acute brain injury. This review discusses how minocycline differs from failed agents, the mechanisms of its acute neuroprotective effect, and the pharmacokinetic and toxicity information necessary to use minocycline for acute neuroprotection in humans.
Background
Minocycline is a widely used semi-synthetic tetracycline antibiotic7 with known anti-inflammatory, anti-apoptotic and glutamate–antagonist properties in several models of brain injury. As an anti-infective agent, it has been used for decades to treat infections caused by a variety of gram-negative and gram-positive organisms. Minocycline is indicated for several diseases including acne vulgaris,8 CNS and UTI infections, gonorrhea, meningitis, shigellosis, conjunctivitis, psittacosis, Q fever, relapsing fever, syphilis and others. Minocycline is a generic drug that has been used in humans as oral or intravenous formulations and subgingival sustained release microspheres (Arestin) are used in adult patients with periodontitis.9 In bacteria, minocycline, as other members of the tetracycline class of compounds, acts by interfering with protein synthesis through binding to the 30S ribosomal subunit, which results in inhibition of mRNA/tRNA interaction and protein translation.10 However, evidence supporting the anti-inflammatory actions of the tetracyclines, aside from the anti-bacterial properties, has emerged in recent years.11,12 Due to its anticollagenase, immunosuppressive and immunomodulating effects, minocycline HCl has been used for the management of rheumatoid arthritis.13
Neuroprotective properties of minocycline
Over the last 5 years, there have been a large number of reports demonstrating the efficacy of minocycline in a variety of animal models of acute neurological injury (Table 1).14–24 These studies demonstrate that minocycline has a broad neuroprotective effect, unrivaled by other agents. Minocycline is effective in animal models of global cerebral ischemia,18,19 focal cerebral ischemia,14,17,25 traumatic brain injury (TBI),23 spinal cord injury (SCI),20–22 and intracerebral hemorrhage (ICH)24. All these injuries share some common pathophysiological mechanisms and the need for ultra-early (probably within 3–6 hours of onset) interventions and treatment. There is not only reduction in tissue injury but there is also improvement in functional recovery with minocycline treatment.
Table 1.
Model | Animal used | Dose | Time window | Outcome |
---|---|---|---|---|
Focal ischemia; TMCAoa (90 minutes) | Rat | 45 mg/kg IPb bid x first day; 22 mg/kg IP bid X 2 subsequent days | Pretreat 2 or 4 hours |
76% infarct reduction (72 hours) 63% reduction14 |
Focal ischemia; Embolic clot model | Rat | 45 mg/kg IP bid X first day 22.5 mg/Kg IP bid X 2nd day |
1 hr | 42% infarct reduction (48 hours)15 |
Focal ischemia; PMCAoc | Mice | 90 mg/Kg IP | Pretreat 60 min before or 30 min Post | Reduction in infarction and brain swelling16 |
Focal ischemia; TMCAo (90 minutes) | Rat | 3 mg/kg IV and 10 mg/kg IV | 4 and 5 hrs | 40–50% infarct reduction (24 hours)17 |
Global ischemia | gerbils | 45 mg/kg IP 90 mg/Kg IP bidX1st day; 45 mg/Kg after 36 hrs |
pretreat 30 minutes post |
Increase survival of CA1 neurons from 10% to 77% Increase to 71%18 |
Neonatal Hypoxia- ischemia (Carotid occlusion+hypoxia) | Rat pups | 45 mg/kg IP or 22.5 mg/Kg IP | Immediately before before or after hypoxic insult | Robust protection with pre and 30 minutes post but not 3 hours post10 |
SCId | Rat | 50mg/kg IP bidX2days | 30 min post | Improved functional outcome20 |
SCI | Rat | 90mg/kg IP;45mg/Kg IP bidX5 Subsequent days | 1 hr post | Enhance long-term hind limb locomotion, coordinated motor function and hind-limb reflex recovery21 |
SCI (Extradural compression with aneurysm clip) | Mouse | 50 mg/kg | 1 hr post | Improved hind limb function and strength; axonal sparing; superior to methylprednisolone22 |
Traumatic brain Injury | Mouse | 90mg/kg IP 45 mg/Kg IP Bid till sacrifice |
pretreat or 30 minutes post | Improved functional outcome(Rotarod); decrease in lesion size23 |
ICHe (Collagenase) | Rat | 45 mg/Kg IP bidX1day; 22.5 mg/Kg IP | 1 hour | Improvement in functional outcome24 |
Temporary middle cerebral artery occlusion
Intra-peritoneal
Permanent middle cerebral artery occlusion
Spinal cord injury
Minocycline is likely to be more successful than other studied neuroprotective compounds in that it avoids all of the common pitfalls stated above. It has been shown to demonstrate superior blood-brain barrier penetration26 and be protective at an extended time window (greater than 3 hours) in experimental animals.14,17,24 In addition, since it is a safe compound, it is particularly well suited to a clinical trial. Minocycline appears to be an ideal candidate to overcome the issues identified in the previous failed neuroprotective trials.
Mechanisms of action of minocycline as neuroprotective agent
Anti-inflammatory effects
The anti-inflammatory actions of tetracyclines have been demonstrated in both acute and chronic brain injury. Minocycline has anti-inflammatory effects on neutrophils, monocytes, microglial cells, and neurons. Minocycline inhibits neutrophil-mediated tissue injury via inhibition of neutrophil migration and degranulation, as well as suppression of oxygen-radical formation.27 In a focal cerebral ischemia model, minocycline inhibits enzymes that contribute to inflammation such as the inducible form of nitric oxide synthase (iNOS) and Interleukin-1beta-Converting Enzyme (ICE-1),18 suppresses apoptosis, and reduces microglial activation.14,18 Minocycline inhibits nitric oxide (NO) release (likely due to suppression of NOS expression) from monocytic cells induced by lipopolysaccharide (LPS) or interferon-gamma expression.28,29 In an acute toxin model of Parkinsons disease, minocycline protected neurons (induced by MPTP, an oxygen radical based mechanism of injury) where inflammation contributes prominently to neuronal injury. In this model, minocycline prevented microglial activation and expression of Interleukin-1 (IL-1) and iNOS.30 In a rat model of immune/inflammatory encephalitis (EAE), where microglia, monocytes, and T-cell activation mediate neuronal injury via multiple inflammatory mediators, minocycline delayed and reduced the progression of the disease (including demyelination) as well as inflammatory cell infiltration.31
Minocycline at nanomolar concentration inhibits glutamate excitotoxic effects in mixed neuron/glia cell culture correlating with inhibition of p38 phosphorylation and IL-1 release.32 Minocycline protects rat neurons (cerebellar granule) from excitotoxic injury induced by reactive oxygen species and NO. Minocycline neuroprotection in vitro is associated with inhibition of inflammatory signaling kinases such as p38.33 In an EAE model, minocycline reduced TNF release from activated oligodendrocytes while enhancing the release of Interleukin-10 (IL-10) (an anti-inflammatory cytokine).31
The anti-inflammatory effects of minocycline have also been demonstrated in humans. At doses commonly used in humans for other indications, minocycline was shown to provide anti-inflammatory benefits in rheumatoid arthritis patients that have not been treated by other disease modifying agents.34,35 Recently, in a small pilot clinical trial in multiple sclerosis, minocycline at a dose of 200 mg daily reduced the number of gadolinium-enhancing lesions on magnetic resonance imaging (MRI)36, demonstrating its ability to decrease the inflammatory damage associated with the disease. In summary, compelling evidence has been raised in the past few years to suggest that minocycline modulates inflammation and this might represent a novel therapeutic approach to diseases that are characterized by elevation of inflammatory cascades, such as in acute ischemic brain injury.
Anti-apoptotic effect
Apoptosis is thought to play a role in both acute and chronic brain injury. Minocycline has been shown to prevent this programmed cell death (apoptosis) and the release of cytochrome c from mitochondria in a number of in vitro and in vivo models. Minocycline delays progression of Amyotrophic Lateral Sclerosis (ALS)- like syndrome in Superoxide dismutase-1 (SOD-1) mutant mice and inhibits mitochondria cytochrome c release (in vitro and in vivo).37 Minocycline inhibits both caspase-dependent (cytochrome c and Smac/Diablo release) and caspase-independent (apoptosis inducing factor release) mitochondrial cell death in a Huntington striatal cell model.38 Minocycline protects renal proximal tubule cells from apoptosis upon exposure to azide, hypoxia, staurosporine, and cisplatinum.39 Minocycline induces the upregulation of the anti-apoptotic protein Bcl-2, at the messenger ribonuclease (mRNA) level. In fact, the anti-apoptotic effects of minocycline are lost when the cells are pretreated with bcl-2 antisense suggesting that the anti-apoptotic action of minocycline is dependent upon bcl-2. Moreover, bcl-2 is also upregulated in neurons in vitro when incubated with equivalent doses of clinically therapeutic concentrations of minocycline.40 In cardiomyocytes exposed to anoxia-reoxygenation, minocycline inhibited the release of cytochrome c and Smac/Diablo from mitochondria and inhibited caspase activation and apoptosis.41
Inhibition of matrix metalloproteinases (MMPs)
The tetracyclines are known inhibitors of the MMPs.42 Low dose doxycycline was the first FDA-approved MMP inhibitor and is used in periodontal disease.43 In a rat model of adjuvant arthritis, doxycycline and tetracycline (two close analogs of minocycline), in combination with a standard non-steroidal anti-inflammatory agent, reduced joint swelling and inflammation and improved radiological evidence of damage. In this model, the arthritic syndrome was associated with suppression of MMP-2 (gelatinase) activation obtained from the inflamed joints.44 Minocycline also reduced MMP-9 in an EAE model.31 In a collagenase-induced ICH model, minocycline reduced MMP-12 and improved functional outcome.24I It has also been shown that minocycline reduces renal microvascular leakage in a rat model of ischemic renal injury (IRI). This action is probably due to diminishing the activity of MMPs.45 MMPs are increasingly being associated with diseases that involve degeneration of extracellular proteins and matrix in the brain46. For that reason, the inhibition of these proteases through minocycline seems to be an attractive experimental therapeutic approach.
Summary of mechanisms of action
Minocycline has multiple mechanisms of action including anti-inflammatory, MMP inhibition and anti-apoptotic that make it an attractive neuroprotective agent. It has activity in multiple acute and chronic animal models of neurological disease. It is one of the few agents that shows efficacy in animal models of spinal cord injury (SCI), traumatic brain injury, intracerebral hemorrhage (ICH), global and focal cerebral ischemia. This points to another key element that distinguishes minocycline from other neuroprotective agents: the diversity of cellular mechanisms affected. Furthermore, it is likely that minocycline may be acting through vascular mechanisms also, which in the past years have been correlated with the intensity of the inflammatory response to injury and severity of damage to the brain parenchyma.47 Finally, in humans, minocycline appears to exert anti-inflammatory properties at the same dosing regimens that are clinically utilized for anti-bacterial treatment. Therefore, minocycline is a candidate drug for neuroprotection in acute brain injury in its present human formulation and dose.
Pharmacokinetic issues
In acute brain injury, the ability to deliver a potential neuroprotective agent rapidly to the systemic circulation is a necessity, most often requiring intravenous administration. Since minocycline has been used for decades, the clinical pharmacokinetics are well - described In humans. After a 200 mg intravenous dose, peak concentrations are, on average, 4.0 mg/L, 48 and steady-state concentrations after 100 mg orally twice daily for 3 days range from 1.4 – 1.8 mg/L.26 Minocycline is the most lipophilic of the commonly used tetracycline antibiotics and cerebral spinal fluid (CSF) concentrations of 11 – 56% of plasma concentrations are achieved.26 Therefore, one would expect CSF concentrations after chronic dosing to be approximately 0.5 mg/L. In addition, minocycline has lower urinary excretion than other tetracyclines, so it is safer to use for patients with renal insufficiency.
Since 1999, most of the published evidence of the neuroprotective effects of minocycline in rodent models of brain injury have employed large intraperitoneal (IP) doses,14,17,19,23,24,25,39,49,50 ranging from a low of 10 mg/kg to a high of 90 mg/kg.16,23 Even in stroke models, where timely delivery of neuroprotection to the brain is known to be important, IP administration was used.14,19,24,39 Since the pharmacokinetics of large IP doses of minocycline in rodents was unknown and necessary in order to translate the experimental research to humans, we performed a study to accomplish this. We found that the IP route resulted in widely variable minocycline serum concentrations and delayed absorption into the systemic circulation, with peak concentrations achieved, on average, 2.5 hours after injection. When compared to intravenous (IV) administration, bioavailability ranged from 10% to 80%, probably due to frank deposition of the drug in the peritoneal cavity.51 The IP route of administration probably accounts for the wide range of high doses in the literature. Use of IV dosing was needed to determine the “true” therapeutic window and dose-response relationship in focal cerebral ischemia.51 We have determined that peak serum concentrations above 3.5 mg/L and trough concentration above 2mg/L are neuroprotective in temporary focal cerebral ischemia in rats.17 Lower doses are currently being tried.
When the absorption problems of both oral and IP administration are overcome by IV administration, the volume of distribution of minocycline is similar in rats and humans when adjusted by weight.51 In other words, administration of 3 mg/kg IV to humans and rats would be expected to achieve peak concentrations of the same magnitude (3–5 mg/L). The main difference in the pharmacokinetic parameters between the two species is the half-life, which is approximately 17 hours in humans26 and only 3 hours in rats.51,52 In summary, intravenous minocycline, in doses commonly used in humans, should achieve serum and CSF concentrations that have been shown to be neuroprotective in animal models.
Adverse effects of minocycline
Most of the information available on the tolerability of minocycline has been obtained after chronic oral administration. In ambulatory patients taking minocycline chronically, raising the dose beyond 100 mg twice daily has been problematic because of the common (26–78% of patients) side effect of dizziness.8,53 More recently, all adverse effects of oral minocycline in ALS patients were found to be gastrointestinal (GI) in nature.54 The average tolerated dose was 387 mg daily and all patients could tolerate at least 300 mg daily. No dizziness was experienced but elevations in blood urea nitrogen and liver enzymes were reported over the 6-month treatment period. Doses of up to 400 mg IV have been used safely for the treatment of serious infections in humans. In a case series of 119 patients receiving from 200–400 mg of intravenous minocycline for 2–24 days for the treatment of infectious diseases,55 21/119 (18%) experienced side effects, 50% of which were gastrointestinal in nature. Only one patient discontinued therapy prematurely and this was a patient who developed azotemia and the attribution was complicated by a case of chronic urinary tract infection. In a search of all reported adverse effects with intravenous minocycline reported to the World Health Organization (WHO) Collaborating Center for International Drug Monitoring, Uppsala, Sweden since 1975, 122 case reports of adverse drug reactions were reported. No assessment of causality was given and the report does not represent the opinion of the WHO. The most common event reported was hepatic function abnormality (19 reports). Thrombocytopenia was reported 11 times and one case of injection site reaction was reported. The WHO data is limited by the fact that no denominator can be ascertained and the dose and duration of intravenous minocycline administered is also unknown.
It is still unknown what dose of minocycline will be neuroprotective and tolerable in human patients. In addition, the feasibility of rapidly administering intravenous doses of minocycline to patients and preliminary evidence of activity of the compound should be determined before committing to further development of minocycline as a therapeutic strategy for acute neuroprotection. The question of optimal duration should be addressed through assessment of a biomarker of inflammation and minocycline serum levels in the patient. In addition, further translational studies in experimental animals will contribute to our understanding of the optimal duration of minocycline treatment in neuroprotection.
Conclusion
Minocycline, which is already in clinical trials for the chronic brain injury of ALS and MS, has a strong therapeutic potential for brain diseases that require acute intervention, such as stroke. It has long been established as a safe drug for clinical use, has multiple mechanisms of action and has a delayed therapeutic window in experimental models. Minocycline is ready for clinical trials as an acute neuroprotectant.
References
- 1.Becker KJ. Targeting the central nervous system inflammatory response in ischemic stroke. Curr Opin Neurol. 2001;14:349–353. doi: 10.1097/00019052-200106000-00014. [DOI] [PubMed] [Google Scholar]
- 2.Martinez-vila E, Sieria PI. Current status and perspectives of neuroprotection in ischemic stroke treatment. Cerebrovasc Dis. 2001;11 (suppl 1):60–70. doi: 10.1159/000049127. [DOI] [PubMed] [Google Scholar]
- 3.Lees KR. Neuroprotection. Br Med Bull. 2000;56:401–412. doi: 10.1258/0007142001903049. [DOI] [PubMed] [Google Scholar]
- 4.DeGraba TJ, Pettigrew LC. Why do neuroprotective drugs work in animals but not humans? Neurol Clin. 2000;18(2):475–493. doi: 10.1016/s0733-8619(05)70203-6. [DOI] [PubMed] [Google Scholar]
- 5.Anonymous. Neuroprotection as initial therapy in acute stroke. Third Report of an Ad Hoc Consensus Group Meeting. The European Ad Hoc Consensus Group. Cerebrovasc Dis. 1998;8(1):59–72. doi: 10.1159/000015817. [DOI] [PubMed] [Google Scholar]
- 6.Gladstone DJ, Black SE, Hakim AM. Toward wisdom from failure: lessons from neuroprotective stroke trials and new therapeutic directions. Stroke. 2002;33:2123–2136. doi: 10.1161/01.str.0000025518.34157.51. [DOI] [PubMed] [Google Scholar]
- 7.Nelson ML. Chemical and biological dynamics of tetracyclines. Adv Dent Res. 1998;12:5–11. doi: 10.1177/08959374980120011901. [DOI] [PubMed] [Google Scholar]
- 8.Coskey RJ. Acne: treatment with minocycline. Cutis. 1976;17(4):799–801. [PubMed] [Google Scholar]
- 9.www.mdconsult.com visited 6–20–2005
- 10.Goodman, Gilman . In: The Pharmacological Basis of Therapeutics. Hardman JG, Limbrid LE, editors. Chapter 47. 2001. pp. 1240–1246. [Google Scholar]
- 11.Golub LM, McNamara TF, d’Angelo GD, Greenwald RA, Ramamurthy NS. A non-antibacterial chemically modified tetracycline inhibits mammalian collagenase activity. J Dent Res. 1987;66:1310–1314. doi: 10.1177/00220345870660080401. [DOI] [PubMed] [Google Scholar]
- 12.Gabler WL, Creamer HR. Suppression of human neutrophil functions by Tetracyclines. J Periodontal Res. 1991;26:52–58. doi: 10.1111/j.1600-0765.1991.tb01626.x. [DOI] [PubMed] [Google Scholar]
- 13.Tilley BC, Alarcon GS, Heyse SP, et al. Minocycline in Rheumatoid Arthritis. A 48-week, double-blinded, placebo-controlled trial. Ann Int Med. 1995;122:81–89. doi: 10.7326/0003-4819-122-2-199501150-00001. [DOI] [PubMed] [Google Scholar]
- 14.Yrjänheikki J, Tikka T, Keinänen R, Goldsteins G. A tetracycline derivative, minocycline, reduces inflammation and protects against focal cerebral ischemic with a wide therapeutic window. Proc Natl Acad Sci USA. 1999;96:13496–13500. doi: 10.1073/pnas.96.23.13496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Wang CX, Yang T, Noor R, Shuaib A. Delayed minocycline but not delayed mild hypothermia protects against embolic stroke. Biomed Cent Neurol. 2002;2:2–8. doi: 10.1186/1471-2377-2-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Morimoto N, Shimazawa M, Yamashima T, Nagabi H, Hara H. Minocycline inhibits oxidative stress and decreases in vitro and in vivo ischemic neuronal damage. Brain Res. 2005;1044:8–15. doi: 10.1016/j.brainres.2005.02.062. [DOI] [PubMed] [Google Scholar]
- 17.Xu L, Fagan SC, Waller JL, et al. Low dose intravenous minocycline is neuroprotective after middle cerebral artery occlusion-reperfusion in rats. BMC Neurology. 2004 Apr 26;4:7. doi: 10.1186/1471-2377-4-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Yrjänheikki J, Keinänen R, Pellikka M, Hokfelt T, Koistinaho J. Tetracyclines inhibit microglial activation and are neuroprotective in global brain ischemia. Proc Natl Acad Sci USA. 1998;95:15769–15774. doi: 10.1073/pnas.95.26.15769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Arvin KL, Han BH, Du Y, Lin S, Paul SM, Holtzman DM. Minocycline markedly protects the neonatal brain against hypoxic-ischemic injury. Ann Neurol. 2002;52:54–61. doi: 10.1002/ana.10242. [DOI] [PubMed] [Google Scholar]
- 20.Stirling DP, Khodarahmi K, Liu J, et al. Minocycline treatment reduces delayed oligodendrocyte death, attenuates axonal dieback, and improve functional outcome after spinal cord injury. J Neurosci. 2004;24:2182–2190. doi: 10.1523/JNEUROSCI.5275-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Teng YD, Choi H, Onario RC, et al. Minocycline inhibits contusion-triggered mitochondrial cytochrome c release and mitigates functional deficits after spinal cord injury. Proc Natl Acad Sci USA. 2004;101:3071–3076. doi: 10.1073/pnas.0306239101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Wells JEA, Hurlbert RJ, Fehlings MG, Yong VW. Neuroprotection by minocycline facilitates significant recovery from spinal cord injury in mice. Brain. 2003;126:1628–1637. doi: 10.1093/brain/awg178. [DOI] [PubMed] [Google Scholar]
- 23.Sanchez Mejia RO, Ona VO, Li M, Friedlander RM. Minocycline reduces traumatic brain injury-mediated caspase-1 activation, tissue damage, and neurological dysfunction. Neurosurg. 2001;48:1393–1401. doi: 10.1097/00006123-200106000-00051. [DOI] [PubMed] [Google Scholar]
- 24.Power C, Henry S, Del Bigio MR, et al. Intracerebral hemorrhage induces macrophage activation and matrix metalloproteinases. Ann Neurol. 2003;53:731–742. doi: 10.1002/ana.10553. [DOI] [PubMed] [Google Scholar]
- 25.Wang CX, Yang T, Schuaib A. Effects of minocycline alone and in combination with mild hypothermia in embolic stroke. Brain Res. 2003;963:327–329. doi: 10.1016/s0006-8993(02)04045-3. [DOI] [PubMed] [Google Scholar]
- 26.Saivin S, Hovin G. Clinical pharmacokinetics of doxycycline and minocycline. Clin Pharmacokinet. 1988;15:355–366. doi: 10.2165/00003088-198815060-00001. [DOI] [PubMed] [Google Scholar]
- 27.Golub LM, Ramamurthy NS, McNamara TF, Greenwald RA, Rifkin BR. Tetracyclines inhibit connective tissue breakdown: new therapeutic implication for an old family of drugs. Crit Rev Oral Biol Med. 1991;2:297–322. doi: 10.1177/10454411910020030201. [DOI] [PubMed] [Google Scholar]
- 28.Amin AR, Attur MG, Thakker GD, et al. A novel mechanism of action of Tetracyclines: Effects of nitric oxide synthases. Pro Natl Acad Sci USA. 1996;93:14014–14019. doi: 10.1073/pnas.93.24.14014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Lee SM, Yune TY, Kim SJ, et al. Minocycline inhibits apoptotic cell death via attenuation of TNF-α expression following iNOS/NO induction by lipopolysaccharide in neuron/glia co-culture. J Neurochem. 2004;91:568–578. doi: 10.1111/j.1471-4159.2004.02780.x. [DOI] [PubMed] [Google Scholar]
- 30.Du Y, Ma Z, Lin S, et al. Minocycline prevents nigrostriatal dopaminergic neurodegeneration in the MPTP model of Parkinsons Disease. Pro Natl Acad Sci USA. 2001;98:14669–14674. doi: 10.1073/pnas.251341998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Brundula V, Rewcastle NB, Metz LM, Bernard CC, Yong VW. Targeting leukocyte MMPs and transmigration: Minocycline as a potential therapy for multiple sclerosis. Brain. 2002;125(Pt 6):1297–1308. doi: 10.1093/brain/awf133. [DOI] [PubMed] [Google Scholar]
- 32.Tikka T, Fiebich BL, Goldsteins G, Keinanen R, Koistinaho J. Minocycline, a tetracycline derivative, is neuroprotective against excitotoxicity by inhibiting activation and proliferation of microglia. J Neurosci. 2001;21:2580–2588. doi: 10.1523/JNEUROSCI.21-08-02580.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Lin S, Zhang Y, Dodel R, Farlow MR, Paul SM, Du Y. Minocycline blocks nitric oxide-induced neurotoxicity by inhibition of p38 MAPK in rat cerebellar granule neurons. Neurosci Lett. 2001;315:61–64. doi: 10.1016/s0304-3940(01)02324-2. [DOI] [PubMed] [Google Scholar]
- 34.Furst DE. Update on clinical trials in the rheumatic diseases. Curr Opin Rheumatol. 1998;10:123–128. doi: 10.1097/00002281-199803000-00007. [DOI] [PubMed] [Google Scholar]
- 35.O’Dell JR, Blakely KW, Mallek JA, et al. Treatment of early seropositive rheumatoid arthritis: a two year double-blind comparison of minocycline and hydroxychloroquine. Arthritis Rheum. 2001;44:2235–2241. doi: 10.1002/1529-0131(200110)44:10<2235::aid-art385>3.0.co;2-a. [DOI] [PubMed] [Google Scholar]
- 36.Metz LM, Zhang Y, Yeung M, et al. Minocycline reduces gadolinium-enhancing magnetic resonance imaging lesions in multiple sclerosis. Ann Neurol. 2004;55:756. doi: 10.1002/ana.20111. [DOI] [PubMed] [Google Scholar]
- 37.Zhu S, Stavrovskaya IG, Drozda M, et al. Minocycline inhibits cytochrome c release and delays progression of amyotrophic lateral sclerosis in mice. Nature. 2002;417:74–78. doi: 10.1038/417074a. [DOI] [PubMed] [Google Scholar]
- 38.Wang X, Zhu S, Drozda M, et al. Minocycline inhibits caspase-independent and –dependent mitochondrial cell death pathways in models of Huntington’s disease. Pro Natl Acad Sci USA. 2003;100:10483–10487. doi: 10.1073/pnas.1832501100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Wang J, Wei Q, Wang C, Hill WD, Hess DC, Dong Z. Minocycline upregulates Bcl-2 and protects against cell death in mitochondria. J Biol Chem. 2004;279:19948–19954. doi: 10.1074/jbc.M313629200. [DOI] [PubMed] [Google Scholar]
- 40.Matsukawa N, Yasuhara T, Xu L, Yu G, Hess DC, Ojika K, Borlongan CV. Minocycline protects neurons, but not astrocytes against oxygen glucose deprivation via apoptotc pathway. Exp Neurol (abstract) 2005;193:253. [Google Scholar]
- 41.Scarabelli TM, Stephanou A, Pasini E, et al. Minocycline inhibits caspace activation and reactivation, increases the ratio of XIAP to smac/Diablo, and reduces the mitochondrial leakage of cytochrome C and smac/Diablo. J Am Coll Cardiol. 2004;43(5):865–74. doi: 10.1016/j.jacc.2003.09.050. [DOI] [PubMed] [Google Scholar]
- 42.Rifkin BR, Vernillo AT, Golub LM. Blocking periodontal disease progression by inhibiting tissue-destructive enzymes: a potential therapeutic role for tetracyclines and their chermically-modified analogs. Periodontol. 1993;64(8 Suppl):819–827. doi: 10.1902/jop.1993.64.8s.819. [DOI] [PubMed] [Google Scholar]
- 43.Ashley RA. Clinical trials of a matrix metalloproteinase inhibitor in human periodontal disease. SDD Clinical Research Team. Ann N Y Acad Sci. 1999;878:335–346. doi: 10.1111/j.1749-6632.1999.tb07693.x. [DOI] [PubMed] [Google Scholar]
- 44.Greenwald RA, Moak SA, Eamamurthy NS, Golub LM. Tetracyclines suppress matrix metalloproteinase activity in adjuvant arthritis and in combination with Flurbiprofen, ameliorate bone damage. J Rheumatol. 1992;19(6):927–38. [PubMed] [Google Scholar]
- 45.Sutton TA, Kelly KJ, Mang HE, Plotkin Z, Sandoval RM, Dagher PC. Minocycline reduces renal microvascular leakage in a rat model of ischemic renal injury. Am J Physiol Renal Physiol. 2005;288:F91–F97. doi: 10.1152/ajprenal.00051.2004. [DOI] [PubMed] [Google Scholar]
- 46.Rosenberg GA, Navratil M, Barone F, Feuerstein G. Proteolytic cascade enzymes increase in focal cerebral ischemia in rat. J Cereb Blood Flow Metab. 2003 Aug;23(8):879–94. doi: 10.1097/00004647-199605000-00002. [DOI] [PubMed] [Google Scholar]
- 47.del Zoppo GJ, Hallenbeck JM. Advances in the vascular pathophysiology of ischemic stroke. Thromb Res. 2000 May 1;98(3):73–81. doi: 10.1016/s0049-3848(00)00218-8. [DOI] [PubMed] [Google Scholar]
- 48.MacDonald H, Kelly RG, Allen S, Noble JF, Kanegis LA. Pharmacokinetic studies on minocycline in man. Clin Pharmacol Ther. 1973;14:852–861. doi: 10.1002/cpt1973145852. [DOI] [PubMed] [Google Scholar]
- 49.He Y, Appel S, Le W. Minocycline inhibits microglial activation and protects nigral cells after 6-hydroxydopamine injection into mouse striatum. Brain Res. 2001;909:187–193. doi: 10.1016/s0006-8993(01)02681-6. [DOI] [PubMed] [Google Scholar]
- 50.Wu DC, Jackson-Lewis V, Vila M, et al. Blockade of microglial activation is neuroprotective in the 1-methyl-4-phenyl- 1,2,3,6-tetrahydropyridine mouse model of Parkinson disease. J Neurosci. 2002;22:1763–1771. doi: 10.1523/JNEUROSCI.22-05-01763.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Fagan SC, Edwards DJ, Borlongan C, Xu L, Arora A, Feuerstein G, Hess DC. Optimal delivery of minocycline to the brain. Implication for human studies of acute neuroprotection. Exp Neurol. 2004;186:248–251. doi: 10.1016/j.expneurol.2003.12.006. [DOI] [PubMed] [Google Scholar]
- 52.Colovic M, Caccia S. Liquid chromatographic determination of minocycline in brain-to-plasma distribution studies in the rat. J Chrom B. 2003;791:337–343. doi: 10.1016/s1570-0232(03)00247-2. [DOI] [PubMed] [Google Scholar]
- 53.Drew TJ, Altman R, Black K, Goldfield M. Minocycline for prophylaxis of infection with Neisseria meningitides: high rate of side effects in recipients. J Infect Dis. 1976;133:194–198. doi: 10.1093/infdis/133.2.194. [DOI] [PubMed] [Google Scholar]
- 54.Gordon PH, Moore DH, Gelinas DF, et al. Placebo controlled Phase I/II studies of minocycline in amyotrophic lateral sclerosis. Neurology. 2004;62:1845–1847. doi: 10.1212/01.wnl.0000125321.92112.7e. [DOI] [PubMed] [Google Scholar]
- 55.Clark BJ, Hodgson CR, Dornbush AC, Hutchinson J. Minocycline administered intravenously: pharmacological activity and clinical experience. Curr Ther Res. 1974;16:865–877. [PubMed] [Google Scholar]