Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Jun 1.
Published in final edited form as: Bone. 2022 Mar 3;159:116377. doi: 10.1016/j.bone.2022.116377

Tetracyclines and bone: unclear actions with potentially lasting effects

Amy J Warner a,b,c, Jessica D Hathaway-Schrader a,b,c, Rena Lubker d,e, Christopher Davies a,f,I, Chad M Novince a,b,c
PMCID: PMC9035080  NIHMSID: NIHMS1786503  PMID: 35248788

Abstract

Tetracyclines are a broad-spectrum class of antibiotics that have unclear actions with potentially lasting effects on bone metabolism. Initially isolated from Streptomyces, tetracycline proved to be an effective treatment for Gram +/− infections. The emergence of resistant bacterial strains commanded the development of later generation agents, including minocycline, doxycycline, tigecycline, sarecycline, omadacycline, and eravacycline. In 1957, it was realized that tetracyclines act as bone fluorochrome labels due to their high affinity for the bone mineral matrix. Over the course of the next decade, researchers discerned that these compounds are retained in the bone matrix at high levels after the termination of antibiotic therapy. Studies during this period provided evidence that tetracyclines could disrupt prenatal and early postnatal skeletal development. Currently, tetracyclines are most commonly prescribed as a long-term systemic therapy for the treatment of acne in healthy adolescents and young adults. Surprisingly, the impact of tetracyclines on physiologic bone modeling/remodeling is largely unknown. This article provides an overview of the pharmacology of tetracycline drugs, summarizes current knowledge about the impact of these agents on skeletal development and homeostasis, and reviews prior work targeting tetracyclines’ effects on bone cell physiology. The need for future research to elucidate unclear effects of tetracyclines on the skeleton is addressed, including drug retention/release mechanisms from the bone matrix, signaling mechanisms at bone cells, the impact of newer third generation tetracycline antibiotics, and the role of the gut-bone axis.

1. Introduction

Tetracyclines are a broad-spectrum class of antibiotics that have unclear actions with potentially lasting effects on bone metabolism (Figure 1). Tetracyclines are used for the treatment of bacterial infections, periodontitis, and dermatological conditions.16 Tetracycline drug-induced bone fluorochrome labeling and minocycline-induced “black bone” discoloration demonstrate that tetracyclines are retained in the bone matrix.711 Tetracyclines have detrimental effects on prenatal and early postnatal skeletal development and can attenuate ovariectomy-induced osteopenia.1220 In vitro investigations have shown that tetracyclines impact osteoclastogenesis and osteoblastogenesis.2129

Figure 1. Tetracyclines and bone.

Figure 1.

[Two-Column Fitting Image] Tetracyclines are used for clinical dynamic bone histomorphometry as well as for the treatment of bacterial infections, periodontitis, and dermatological conditions. Tetracycline antibiotic bone fluorochrome labeling and minocycline-induced black bone discoloration demonstrate that tetracyclines are retained in the bone matrix. Tetracyclines have detrimental effects on early skeletal development and can attenuate experimentally induced osteopenia. In vitro investigations have delineated that tetracyclines can suppress osteoclastogenesis and suppress or promote osteoblastogenesis.

2. Pharmacology of Commonly Administered Tetracyclines

Tetracyclines are a class of antibiotics that were first discovered in 1948 when Duggar isolated chlortetracycline from the soil bacterium Streptomyces aureofaciens.30 Soon thereafter, other natural tetracyclines produced by Streptomyces were also isolated, including oxytetracycline and tetracycline.2 Tetracycline was favored clinically due to its higher potency, better solubility profile and more favorable pharmacological activity.1

The tetracycline scaffold is based on a ABCD naphthacene ring system containing four aromatic rings (Figure 2A). Tetracycline antibiotics exert their antimicrobial activity through inhibition of bacterial protein synthesis by binding to the decoding center in the 30S ribosomal subunit, thus preventing the binding of aminoacyl-tRNA to the A site. Specifically, the conserved hydrophilic surface of tetracycline interacts with the irregular minor groove of helix 34 and loop of helix 31 of 16S rRNA.31 In this position, the C and D rings of tetracycline (Figure 2B) sterically hinder the interaction between the first nucleotide of the anticodon of the tRNA and the third base of the mRNA codon.31 Modifications to the ABCD naphthacene ring system alter the pharmacological profiles of tetracycline drugs.

Figure 2. Chemical structure of tetracycline drugs.

Figure 2.

(A) ABCD napthacene core ring system (B) Naturally occurring tetracycline. (C) Doxycycline, an analog of tetracycline, with a rearrangement of the methyl group from position carbon 5 (C5) to position C6. (D) Minocycline, an analog of tetracycline, with a dimethylamino group at position C7 and lacking the methyl and hydroxy groups at position C5. (E) Tigecycline, an analog of minocycline, with a 9-tert-butyl-glycylamido side chain at position C9 on the D ring. (F) Omadacycline, an aminomethylcycline with structural modifications of minocycline at D9 with an aminomethyl group. (G) Eravacycline contains a tetracyclic core, with the addition of a fluorine at position C7 and a pyrrolidinoacetamo group at C9. (H) Sarecycline, another minocycline analog, but with a replacement of the dimethylamino group at position C.

The emergence of antimicrobial resistance to early tetracyclines spurred the development of later generation tetracycline drugs.32,33 Doxycycline and minocycline are second generation, semisynthetic tetracycline antibiotics introduced in the 1960s.1,34 Doxycycline (Figure 2C) was designed to increase drug stability, as well as oral availability with improved lipid solubility, by shifting the methyl group on tetracycline from ring B to ring C.35,36 Compared to its tetracycline parent, minocycline (Figure 2D) contains an additional dimethylamido group on carbon 7 (C7) of the D ring and exhibits increased affinity to the rRNA target, through increased stacking interactions with helix 34.31 Tigecycline (Figure 2E), a third generation semisynthetic tetracycline drug, was introduced in 2005. Tigecycline exhibits a chemical modification to minocycline at C9 on the D ring.3739 Introduction of a 9-tert-butyl-glycylamido side chain enhances binding affinity by increasing stacking interactions with 16S rRNA.31,40 In 2018, the FDA approved three new tetracycline drugs, including two semisynthetic agents, omadacycline and sarecycline, and the first fully synthetic compound, eravacycline.41 Omadacycline (Figure 2F) is a minocycline derivative with a C9 substitution on the D ring. The aminomethyl group substitution on ring D confers 2-fold higher affinity for the ribosome and inhibits in vitro translation at 2-fold lower drug concentrations compared to tetracycline.31 In addition, the C9 substitution increases oral bioavailability and lowers the adverse gastrointestinal effects associated with tigecycline.40 Eravacycline (Figure 2G) contains a tetracycline core with modifications that include a fluorine atom at C7 and a pyrrolidinoacetamido group at C9 of the D ring. This confers a 10-fold higher affinity for the ribosome and inhibits in vitro translation at 4-fold lower drug concentrations compared to tetracycline.31 Sarecycline (Figure 2H) is a minocycline analog that has 7-[[methoxy(methyl)amino]methyl] group introduction at C7 on the D ring. The C7 moiety interacts directly with mRNA at the A-site codon. This interaction may be responsible for sarecycline’s reduced adverse side effects compared to other tetracycline drugs.42

3. Clinical Applications of Tetracyclines

Tetracycline, doxycycline, minocycline, and tigecycline are the most commonly used tetracycline antibiotics when treating bacterial infections (Table 1)43,44. Tigecycline use is limited to resistant infections that are refractory to earlier generation tetracyclines. Tigecycline use is restricted because an increase in all-cause mortality was observed among adult patients during clinical trials of the agent.45,46 Omadacycline was introduced to treat adults with community-acquired bacterial pneumonia and acute bacterial skin and skin structure infections, including those resistant to earlier generation tetracyclines.41,47 Eravacycline is used to treat complicated, antibiotic resistant intra-abdominal infections.48,49 Sarecycline is FDA approved for the treatment of moderate to severe acne41 and was developed to limit adverse gastrointestinal and phototoxicity side effects caused by off-target actions of earlier generation tetracycline drugs.50 The low incidence of adverse gastrointestinal effects with sarecycline is due to its reduced activity against Gram-negative bacteria and anaerobes present in the intestinal microbiota.50

Table 1:

Clinical uses of tetracycline drugs

Antibiotic Origin Half-life elimination Uses
Tetracycline Natural51 6–11 hours52 Acne vulgaris53, sexually transmitted infections54, Vibrio cholerae55
Off-label·. Helicobacter pylori eradication56, periodontitis57
Minocycline Semi-synthetic51 11–17 hours58 Acne vulgaris, Lyme disease, perioral infections
Off-label: Gastrointestinal, sexually transmitted, soft tissue, Rickettsia, zoonotic infections59
Doxycycline Semi-synthetic51 18–22 hours60 Acne vulgaris53, periodontitis57, sexually transmitted infections54
Off-label: Anthrax61, malaria62
Tigecycline Semi-synthetic51 ~40 hours63 Complicated skin and intra-abdominal infections64,65
Multidrug-resistant organisms37
Omadacycline Semi-synthetic41 ~16 hours47 Community-acquired bacterial pneumonia, Skin and skin structure infections47
Eravacycline Synthetic41 20 hours48 Complicated intra-abdominal infections41
Sarecycline Semi-synthetic66 21–22 hours67 Acne vulgaris41

In addition to bacterial infections, tetracyclines are also administered to treat dermatological conditions and periodontal disease.1 Of these, the most common use of tetracycline drugs is for the treatment of acne vulgaris.43,68,69 Acne afflicts approximately 50 million Americans annually, of whom roughly 85% are between the ages of 12 and 24.70,71 Overall, about 1/3 of adolescents and young adults are prescribed oral antibiotics for the treatment of acne.72,73 According to the 2016 Status Report from the Scientific Panel on Antibiotic Use in Dermatology of the American Acne and Rosacea Society, tetracyclines constitute 75% of oral antibiotics prescribed by dermatologists.74 The pathophysiology of acne is mostly attributed to the opportunistic bacteria Cutinebacterium acnes (C. acnes), formerly known as Propionibacterium acnes.70,71 Minocycline and doxycycline are administered to target C. acnes7577. Clinical trials have found that sarecycline has similar efficacy against C. acnes, but with reduced adverse gastrointestinal and phototoxicity side effects.7881 Although practice guidelines recommend a minimum of 6–8 weeks and a maximum of 3–6 months for administration of oral antibiotics for acne8284, more than 60% of oral antibiotic courses extend longer than three months, with eleven months being the average duration of treatment.72,73 The standardized doses of oral tetracycline, doxycycline, minocycline, and sarecycline for the treatment of acne in children, adolescents, and adults are summarized in Table 2.

Table 2:

Standard oral dosing of tetracyclines for acne

Antibiotic *Children ≥ 8 yro and adolescents Adults
Tetracycline 500 mg twice daily44 1 g daily in divided doses; reduce gradually to 125 to 500 mg/day53
Doxycycline 50 to 100 mg once or twice daily or 150 mg once daily44 50 to 100 mg twice daily or 100 mg once daily53,85
Minocycline 50 to 100 mg once or twice daily44 50 to 100 mg twice daily53
Sarecycline 60 mg daily (33–54 kg patients)
100 mg daily (55–84 kg patients)
150 mg daily (85–136 kg patients)41
 *Children ≥ 9 yro and adolescents
60 mg daily (33–54 kg patients)
100 mg daily (55–84 kg patients)
150 mg daily (85–136 kg patients)41

Administering tetracyclines at antimicrobial doses (Table 2) can result in a range of undesirable side-effects, including adverse gastrointestinal and hepatic reactions.67,86 Dose-related gastrointestinal side effects, including abdominal discomfort, epigastric pain, nausea, vomiting, and diarrhea, are commonly reported by patients taking oral tetracyclines and intravenous tigecycline.67 Minocycline has most frequently been linked drug-induced liver injury. Short-term use of minocycline may result in acute-hepatitis-like syndrome, while long-term use may cause chronic hepatitis with autoimmune features. These side-effects are associated with a hepatocellular pattern of elevated serum enzymes, autoantibodies, and immunological features such as fever, rash, and eosinophilia.86 Although doxycycline and minocycline share a similar chemical structure, doxycycline is not linked to autoimmune-like hepatitis. Doxycycline-induced liver injury is commonly characterized as a mix between hepatocellular and cholestatic.86 Tetracycline-induced liver injury is rare. Acute fatty liver disease has been reported in pregnant women when given tetracycline in high doses.86 The hepatotoxicity caused by minocycline, doxycycline, and tetracycline typically resolves after terminating antibiotic use.67,86

To reduce these adverse effects, tetracyclines have been administered at sub-antimicrobial dosing for acne treatment.87 In 1998, the FDA approved Periostat (sub-antimicrobial 20 mg doxycycline hyclate capsule administered twice daily) as an adjunctive host immunomodulatory treatment for chronic periodontitis.6,8890 In 2006, the FDA approved Oracea as a sub-antimicrobial dose doxycycline treatment for rosacea, which is administered once daily.91 Oracea is a 40 mg modified-release doxycycline capsule, which consists of 30 mg immediate-release and a 10 mg delayed-release mechanism.3,9294

4. Retention of Tetracyclines in Bone

One of the more remarkable properties of tetracyclines is their retention in the bone matrix. This was first recorded in 1956 when it was observed that a single intravenous injection of tetracycline led to incorporation and retention of the antibiotic in the bone of young mice.7 Later, Milch et al. (1957) administered a single intraperitoneal injection of tetracycline, chlortetracycline, or oxytetracycline (0.1 to 200 mg/kg) to mice, rats, guinea pigs, rabbits, and dogs and observed a brilliant yellow-gold fluorescence under ultraviolet light in long bones and flat bones across all species.8 Fluorescence could be detected at low doses (0.3 mg/kg) and throughout the 10-week observation period.8 Milch et al. (1958) employed subcutaneous, intramuscular, and intravenous injections or oral administration to show that the mode of administration had no effect on tetracycline-induced bone fluorescence in both laboratory animals and human subjects.9 Administering a clinical dose of tetracycline (1–2 g/day) was sufficient to induce bone fluorescence in humans. Whereas sex had no effect, tetracycline-induced bone fluorescence was influenced by age. Labeling was more intense in bone specimens from young versus mature adult subjects and appeared to occur only at regions of active bone formation. Fluorescence was intensely localized to the trabecular bone secondary spongiosa and the endosteal surfaces, periosteal surfaces and haversian canals of cortical bone. Since tetracyclines chelate metallic ions, including calcium, the authors speculated that fluorescence resulted from binding of the tetracycline naphthacenecarboxamide nucleus to calcium in the bone. The authors highlighted the need for future research to elucidate the mechanism by which tetracyclines are incorporated in bone, to measure the half-life when bound to bone, and understand the biological implications for skeletal health.9

To address these questions, subsequent studies evaluated the metabolism of tetracycline, and determined the concentration and persistence of the drug retained in bone.10,95 Kelly and Buyske (1960) and Buyske et al. (1960) showed about 90% of tetracycline was eliminated via the fecal or urinary route, while 3–6 % of the drug was retained by the skeleton in Sherman rats.10,95 Tetracycline was retained in the femur after a single 250 mg/kg dose by intraperitoneal injection and did not begin to deplete until 5–7 months post administration. Prolonged treatment increases the retention of tetracycline in the skeleton, suggesting that bone could act as a drug “depot” for tetracyclines with potentially lasting effects.10 Myers and Jaffe (1965) also demonstrated that tetracycline is retained in the skeleton of Long-Evans rats, with up to 5% of tetracycline retained within the skeleton. An additional finding was that more tetracycline was retained in the bones of young versus mature rats.11

Retention in bone led Frost and collaborators at the Henry Ford Hospital to pioneer the clinical use of tetracycline as a fluorochrome marker for bone.96 Their work showed that independent of the route of administration, tetracycline was deposited in the hydroxyapatite of actively mineralizing bone, hyaline cartilage, and developing teeth.96 Frost and colleagues utilized pulsed tetracycline labeling of bone to develop histomorphometric approaches to quantify physiologic rates of bone formation, mineralization, and remodeling.97100 These approaches were seminal in establishing standardized dynamic bone histomorphometry techniques,101,102 that continue to be used in research and clinical care today. Tetracycline and doxycycline remain the most commonly used bone fluorochrome labels in humans.103

Different from other tetracyclines, minocycline does not have fluorescent properties in bone.103 However, minocycline has been associated with osseous tissue discoloration, providing indirect evidence of retention in the bone matrix.104,105 This phenomenon of minocycline-induced bone discoloration is commonly referred to as “black bone disease”.106109 Case reports in the dental literature commonly document that patients with a history of long-term minocycline therapy present with black/blue pigmentation of the mucogingival tissues.105,109115. Surgical exploration has revealed that alveolar mucogingival tissues were not pigmented, but rather it was the underlying alveolar bone.105,110 Minocycline-induced black/blue bone discoloration has also been observed at non-oral skeletal sites, including the clavicle, vertebrae, acromion, femur, tibia, metatarsals, and metacarpals. In all cases, patients had undergone long-term minocycline therapy for the treatment of acne or rosacea, spanning from 2 to 10 years.104,106108,116121 It is theorized that black/blue discoloration of bone is caused by the oxidation of minocycline and the subsequent incorporation into ossifying tissue.104,110,112,122 Patients are advised to discontinue minocycline treatment if they observe dark pigmentation of the oral mucosa, gingiva, and skin. However, terminating minocycline administration does not resolve the soft tissue pigmentation when the subjacent bone is discolored.112,121 The impact of long-term minocycline retention on bone quality and remodeling is largely unknown.

5. Effects of Tetracyclines on Perinatal and Early Postnatal Skeletal Developmental

Since tetracyclines have affinity for the bone matrix, questions arose whether this would affect early skeletal development (Table 3). Bevelander et al. (1959, 1960) carried out a series of seminal studies evaluating the teratogenic effects of tetracycline by injecting the antibiotic into the yolk sac of eight-day old chick embryos. Whereas lower doses (0.1 to 0.5 mg) had only minimal effects, higher doses (2.5 to 5.0 mg) induced pronounced retardation in the overall growth of the embryos. Moreover, tetracycline stunted skeletal development and led to malformed bones, which was attributed to lower trabeculae numbers and reduced mineralization.12,13 Hughes et al. (1965) later found that injection of chick embryos with a clinically relevant dose of tetracycline (2.5 μg/g) did not interfere with bone growth. Only at ten times higher doses (25 μg/g) was bone growth inhibition and bone malformation observed.123

Table 3:

Tetracycline treatment effects on early skeletal development

Study Model Subject Age Antibiotic Dose Administration Mode Treatment Duration Outcomes
Bevelander et al. (1959,60)12,13 Chick embryo 8 days 2.5 or 5.0 mg Yolk-sac injection Single dose
  • Stunted, malformed bones

  • ↓ Femoral trabeculae & mineralization

Hughes et al. (1965)123 Chick embryo 4 days 25 μg/g Allantois injection Single dose
  • ↓ Bone growth

Bevelander & Cohlan (1962)14 Pregnant rats to assess fetal outcomes - 40–80 mg/kg/day Intramuscular injection 12–20, 8–15, or 10–15 gestation days
  • ↓ Skeletal growth

Cohlan et al. (1963)15 Premature infants 1 to 36 days 14–50 mg/kg/day Oral 9–30 days (4 groups with alternating durations and recovery periods)
  • ↓ Bone growth

Porter et al. (1965)17 Pregnant women to assess postnatal outcomes - 1 g/day Oral 2 or 6 weeks, during 1st, 2nd or 3rd trimester
  • Yellow/brown discoloration and enamel hypoplasia in deciduous dentition

Genot et al. (1970)18 Pregnant women to assess postnatal outcomes - 1 g/day Oral 2 or 6 weeks, during 1st, 2nd or 3rd trimester
  • Yellow/brown discoloration and enamel hypoplasia in deciduous dentition

Wallman & Hilton (1962)131 Healthy Infants 1 day 21–29 mg/kg/day Oral 3 to 6 days
  • Yellow/brown discoloration in deciduous dentition

There is also evidence that tetracyclines affect the skeletal development of mammals in utero. This is of particular concern because antibiotics, including tetracyclines, can traverse the placental barrier from mother to fetus during pregnancy.124126 To ascertain whether transplacental tetracycline impacts the early developing skeleton, Bevelander & Cohlan (1962) administered tetracycline to pregnant rats and found that the skeleton of embryos and newborn pups retained tetracycline and exhibited growth retardation.14 Tetracycline-induced skeletal fluorescence was observed in a nonviable premature infant, whose mother had been administered 1 gram of oral tetracycline daily for 3 weeks prior to delivery.15 This case demonstrated that tetracycline can pass through the placenta and be retained in the human fetal skeleton. To investigate how tetracycline affects fetal skeletal development, Cohlan et al. (1963) administered the antibiotic orally to healthy premature infants.15 Using an x-ray technique to measure fibula growth rate, it was shown that tetracycline caused a dose-dependent inhibition of longitudinal bone growth (14–50 mg/kg/day) by up to 40%. Inhibitory effects on bone growth were reversible in groups in which antibiotic therapy was discontinued after a short period of administration.15 Two double-blind, placebo-controlled clinical studies published in 196517 and 197018 reported the impact of prenatal tetracycline exposure on skeletal growth and development. Bone development was similar in children whose mothers had taken tetracycline during pregnancy in comparison to those who had not. However, tetracycline exposure during the second and third trimesters caused high incidence of yellow/brown discoloration and enamel hypoplasia in the deciduous teeth.17,18 These findings support the view that short-term use of tetracycline during pregnancy poses minimal risk for fetal skeletal development, but that long-term use is contraindicated.127,128 As a result, tetracyclines have been classified as category D drugs. This means that there is evidence of risk to the human fetus, but that benefits from use in pregnant women may outweigh the risk. For this reason, tetracyclines are not recommended as a first-line antimicrobial treatment for pregnant women.67,129

There is also evidence that tetracycline can be excreted in breast milk from mother to offspring. For instance, administering tetracycline to lactating female rats for three weeks following birth resulted in tetracycline labeling in the teeth and skeletons of their offspring.124 As tetracyclines are lipid-soluble drugs, they can diffuse readily through lipid-filled membranes such as those in the mammary glands. Although there have been no reports of adverse effects to nursing infants, clinical studies have shown that tetracyclines are present at low concentrations in the breast milk of mothers receiving tetracycline therapy post-partum.130

The risk of tetracyclines given directly to children must also be considered. Wallman and Hilton (1962) examined 50 children who had been given a course of tetracycline following birth.131 Typically, the tetracycline treatment occurred during the first week of postnatal life. Forty-six of 50 children had yellow/brown discoloration of the deciduous dentition, a characteristic of tetracycline-induced tooth staining.131 Furthermore, studies have shown that administering tetracycline to pediatric patients younger than eight years old results in high incidence of tetracycline-induced tooth staining. This occurs because the mineralization of the succedaneous dentition does not reach completion until a child reaches eight years of age (excluding third molars).132,133 For this reason, clinical guidelines advise that tetracyclines should not be administered to children younger than eight years old.67

6. Impact of Tetracyclines on Osteopenia

6.1. Post-menopausal osteopenia

Tetracycline drugs affinity for the bone matrix further led investigators to study these agents as a therapeutic intervention for post-menopausal bone loss (Table 4). Utilizing the ovariectomized (OVX) rat model, Williams et al. (1996) showed that oral minocycline administration (10 mg/day) improved bone mineral density and trabecular bone microarchitecture properties of the femur. The protective effect of minocycline against osteopenia was attributed to an increase in bone formation, with concurrent decrease in bone resorption.16 Li et al. (2003) found that oral administration of tetracycline (1.2 or 4.8 mg/kg/day) decreased bone resorption and increased osteogenesis in OVX rats. Interestingly, the 1.2 mg/kg/day dose increased osteoblast recruitment, while the higher dose enhanced osteoblast activity.19 Pytlik et al. (2004) later found that doxycycline (20 mg/kg/day) improved OVX-induced impaired bone mechanical properties,20 and de Figueiredo et al. (2019) showed that doxycycline treatment (10 or 30 mg/kg/day) ameliorated OVX-induced osteopenia by improving cancellous bone microarchitecture, increasing bone mineral density, and reducing osteoclastogenesis.134 Together, these findings indicate that tetracyclines have a protective effect on the skeleton in the post-menopausal state.

Table 4:

Tetracycline, minocycline, and doxycycline treatment effects on osteopenia

Study Model Sex Experimental Osteopenia Antibiotic (Dose) Administration Mode Treatment Duration Outcomes
Williams et al. (1996)16 Wister rats F OVX at age 22–24 months Minocycline (10 mg/d) Oral gavage 8 weeks, initiated at OVX
  • ↑ Femur bone mineral density

  • ↑ Femur trabecular bone area, number, and thickness

  • ↑ Bone formation

  • ↓ Bone resorption

Li et al. (2003)19 Sprague Dawley rats F OVX at age 3.5 months Tetracycline (1.2 or 4.8 mg/kg/d) Chow 90 days, initiated at OVX
  • ↑ Trabecular bone area

  • ↑ Osteogenesis

  • ↓ Bone resorption

Pytlik et al. (2004)20 Wister rats F OVX at age 3 months Doxycycline (20 mg/kg/d) Oral gavage 4 weeks, initiated 3–4 days after OVX
  • ↑ Bone mineral content / bone mass ratio in femur

  • Improved mechanical properties of the femur

de Figueiredo et al. (2019)134 Wister rats F OVX at age 2.5 months Doxycycline (10 or 30 mg/kg/d) Drinking water 60 days, initiated 3 months after OVX
  • ↑ Bone mineral density in femur induced by 10 mg/kg/d

  • ↑ Trabecular bone in femur induced by 10 & 30 mg/kg/d

  • ↓ Osteoclasts induced by 30 mg/kg/d

Golub et al. (1990)135 Sprague Dawley rats M STZ-induced diabetes in adult rats Doxycycline (2 mg/d) Oral gavage 6 or 14 weeks
  • ↑ Femur mineral content (calcium, phosphorus)

  • ↓ Urinary calcium excretion

Fowlkes et al. (2015)136 DBA/2J mice M STZ-induced diabetes at age 10–11 weeks Doxycycline (28–92 mg/kg/d) Chow 10 weeks, initiated at age 12–13 weeks
  • No effect on femur trabecular / cortical bone microarchitecture

  • No effect on femur biomechanical properties

Bain et al. (1997)137 Sprague Dawley rats M STZ-induced diabetes at age 3 months Minocycline (20 mg/d) Oral gavage 26 days
  • ↑ Trabecular bone area in tibia

  • ↑ Bone formation and mineral apposition rates in tibia

Sasaki et al. (1991,1992)
138,139
Sprague Dawley rats M STZ-induced diabetes at age 4 months Minocycline (20 mg/d) Oral gavage 21 days
  • ↑ Osteoblast morphology and function

Kaneko et al. (1990)140 Sprague Dawley rats M STZ-induced diabetes in adult rats Minocycline (20 mg/d) Oral gavage 21 days
  • No effect on osteoclast structure and function

6.2. Diabetes-induced osteopenia

Similarly, tetracyclines’ chelating properties in bone led researchers to evaluate the drugs as a therapeutic intervention for diabetes-induced osteopenia (Table 4). In an observational study utilizing doxycycline, Golub et al. (1990) reported that oral doxycycline administration appeared to prevent osteopenia in streptozotocin (STZ)-induced diabetic rats (2 mg/day).135 Contrasting these findings, Fowlkes et al. (2015) observed that orally administering doxycycline (28–92 mg/kg/day) to diabetic DBA/2J mice did not prevent or alleviate diabetes-induced deleterious changes in bone microarchitecture or biomechanical properties.136 On the other hand, Bain et al. (1997) reported that oral administration of minocycline (20 mg/day) prevented cancellous bone loss in STZ-induced diabetic rats. The protective effect against diabetes-induced osteopenia was attributed to improved bone formation and mineral apposition.137 In line with the histomorphometric findings reported by Bain et al. (1997)137, Sasaki and co-authors (1991, 1992) found that orally administering minocycline (20 mg/day) to STZ-induced diabetic rats improved osteoblast morphology and function138,139 while Kaneko et al. (1990) reported that orally administering minocycline (20 mg/day) did not normalize osteoclast outcomes.140 Together, these reports demonstrate there is no clear consensus about tetracyclines’ protective effects on diabetes-induced osteopenia.

7. Impact of Tetracyclines on Osteoclastogenesis

Calcium signaling promotes osteoclast differentiation and function. Recognizing that tetracycline antibiotics chelate calcium, early osteoclast researchers evaluated the effect of tetracyclines on cytosolic calcium (Table 5). Donahue et al. (1992) investigated whether minocycline or doxycycline could alter the levels of cytosolic calcium (Ca2+) in rat osteoclasts in vitro.141 Neither minocycline nor doxycycline (10 μg/ml) altered the cytosolic Ca2+ concentration in osteoclasts. By contrast, Bax et al. (1993) reported that stimulating rat osteoclasts in culture with 10 or 100 mg/ml minocycline increased the concentration of cytosolic Ca2+ by approximately 3X fold.21 Furthermore, Bax et al. (1993) reported that preincubation with minocycline (1 mg/l), but not doxycycline (1 or 10 mg/l), significantly attenuated the cytosolic Ca2+ response in osteoclasts to stimulation with ionic nickel (a surrogate Ca2+ receptor agonist). This led the authors to conclude that minocycline attenuates cytosolic Ca2+ responses in osteoclasts, which is associated with Ca2+ receptor activation.21 Zaidi et al. (1993) reported that minocycline (10 mg/l) stimulation exerted morphometric changes in cultured rat osteoclasts, which were comparable to changes elicited by occupancy of the Ca2+ receptor.22 Overall, these investigations suggest that tetracycline-calcium interactions can influence osteoclastogenesis.

Table 5:

Tetracycline class antibiotic effects on in vitro osteoclastogenesis

Study Cell Culture System Antibiotic Dose Treatment Duration Outcomes
Donahue et al. (1992)141 Primary rat neonatal long bone derived adherent cells Minocycline or doxycycline 10 μg/ml 500 seconds
  • No effect on cytosolic calcium

Bax et al. (1993)21 Primary rat neonatal long bone derived adherent cells Minocycline or doxycycline 0.1, 1, 10, 100 mg/l 1 hour
  • ↑ Cytosolic calcium induced by minocycline

Zaidi et al. (1993)22 Primary rat neonatal long bone derived adherent cells Minocycline 10 mg/l 80 minutes
  • Altered cell morphology

Gomes et al. (1984)142 Rat embryonic bone explant cultures treated with 1.0 μg/ml parathyroid hormone Tetracycline, minocycline, or doxycycline 0.2, 2.0, 20, 200 μg/ml 2 or 5 days
  • ↑ Ca2+ release induced by 20 & 200 μg/ml tetracycline, minocycline, or doxycycline

Zhou et al. (2010)143 Primary rat bone marrow macrophages cultured on bovine bone with 30 ng/ml MCSF, 50 ng/ml RANKL, & 10−7M 1α,25[OH]2D3 Tetracycline 5, 10, 20 μg/ml Apoptosis: 1 day
Formation: 7 days
Resorption: 14 days
  • ↑ Osteoclast apoptosis

  • ↓ Osteoclast formation

  • ↓ Bone resorption

Chowdhury et al. (1993)144 Primary chick embryo long bone derived adherent cells cultured on bovine bone Doxycycline 5 or 15 μg/ml 3, 6, 24 hours
  • ↓ Bone resorption

Bettany et al. (2000)145 Primary murine osteoblasts/osteoclasts co-cultured on ivory wafers Doxycycline 5 or 15 μg/ml 24 hours
  • ↑ Osteoclast apoptosis

  • ↓ Bone resorption

Holmes et al. (2004)23 Primary human peripheral blood mononuclear cells cultured with 30 ng/ml MCSF & 25 ng/ml RANKL Doxycycline 2.5 or 20 μg/ml 20 days
  • ↓ Osteoclast formation induced by 2.5 μg/ml

  • ↑ Osteoclast apoptosis induced by 20 μg/ml

Franco et al. (2011)146 Primary murine bone marrow macrophage cells (BMMCs) stimulated with 10 ng/ml MCSF & 50 ng/ml RANKL
RAW264.7 murine macrophage cells stimulated with 50 ng/ml RANKL
Tetracycline, doxycycline, or minocycline 0.2 or 2.0 μg/ml BMMCs: 6 days
RAW Cells: 5 days
  • ↓ Osteoclast numbers induced by 0.2 & 2.0 μg/ml doxycycline, 0.2 & 2.0 μg/ml minocycline, or 2.0 μg/ml tetracycline

Nagasawa et al. (2011)24 Primary murine bone marrow cells stimulated with 10 nM 1α,25[OH]2D3 or 20 ng/ml RANKL Tetracycline, doxycycline, or minocycline 2.5, 5, 10 μM Formation: 7 days
Viability: 24 hours
  • ↓ Osteoclast formation induced by 2.5, 5, & 10 μM tetracycline; 2.5, 5, & 10 μM doxycycline; or 5 & 10 μM minocycline

  • No effect on cell viability

Kim et al. (2019)25 Primary murine bone marrow macrophage cells stimulated with 30 ng/ml MCSF & 100 ng/ml RANKL Minocycline or tigecycline 1, 2.5, 5 μM 3 days
  • ↓ Osteoclast formation induced by 1, 2.5, & 5 μM minocycline or 1, 2.5, & 5 μM tigecycline

Studies have also shown that tetracycline and its derivatives can have negative effects on osteoclast formation and resorptive function (Table 5). Gomes et al. (1984) assessed the effects of tetracycline, minocycline, and doxycycline (0.2–200 μg/ml) on bone resorption in parathyroid hormone-induced bone organ cultures. Bone resorption was measured by radiolabeled calcium release from embryonic bones. Tetracycline, minocycline and doxycycline had no effect at lower concentrations (0.2 or 2.0 μg/ml), but inhibited bone resorption at higher concentrations (20 or 200 μg/ml).142 Zhou et al. (2010) investigated the effects of tetracycline on osteoclastogenesis in rat bone marrow macrophages stimulated with MCSF and RANKL, which are critical factors for inducing in vitro osteoclastogenesis. Tetracycline at 5 μg/ml did not influence osteoclast formation, resorption, or apoptosis. However, tetracycline inhibited osteoclast formation, attenuated resorptive capacity, and induced apoptosis at concentrations from 10–20 μg/ml.143 Chowdhury et al. (1993) cultured osteoclasts isolated from chick embryo long bones on devitalized bovine cortical bone slices to evaluate the impact of doxycycline on osteoclast function. The addition of doxycycline to the culture system reduced the osteoclast resorptive capacity. Five μg/ml doxycycline reduced the number of resorptive pits by about 50%, and 15 μg/ml doxycycline largely prevented pit formation.144 By co-culturing murine osteoblasts / osteoclasts on ivory wafers, Bettany et al. (2000) similarly demonstrated that doxycycline inhibits osteoclast resorptive activity in vitro. Five μg/ml doxycycline decreased the area resorbed by roughly 50%, and 15 μg/ml doxycycline prevented bone resorption altogether.145 Holmes et al. (2004) investigated doxycycline effects in human peripheral blood mononuclear cell-derived osteoclasts. Mononuclear cells were isolated from the peripheral blood and stimulated with MCSF and RANKL. Doxycycline had no effect on osteoclast differentiation at concentrations less than 250 ng/ml, but reduced osteoclast formation by 50% at 2.5 μg/ml. Doxycycline treatment caused apoptosis in cultured human osteoclasts when concentrations exceeded 20 μg/ml.23 These in vitro studies support the notion that tetracyclines have anti-osteoclastogenic effects.

Subsequent studies investigated the mechanism by which tetracyclines attenuate osteoclastogenesis (Table 5). Franco et al. (2011) evaluated the effects of tetracycline, minocycline, and doxycycline on RANKL-induced RAW264.7 cell osteoclast cultures. Osteoclast numbers per well were reduced by 0.2 and 2.0 μg/ml minocycline, 0.2 and 2.0 μg/ml doxycycline, and 2.0 μg/ml tetracycline.146 The authors found that doxycycline downregulated RANKL-induced osteoclastogenesis through inhibitory effects on RANKL-induced MMP-9 enzyme activity. The inhibitory effects were independent of the transcription factor, NFATc1, which is critical for osteoclast differentiation.146 Nagasawa et al. (2011) evaluated the effects of tetracycline, doxycycline, and minocycline on murine bone marrow cell cultures. Osteoclastogenesis was induced in the culture system via 1α,25[OH]2D3 or RANKL. Tetracycline drug concentrations at 2.5 / 5 / 10 μM inhibited osteoclast formation in a dose-dependent manner. The authors concluded that minocycline inhibits RANKL-induced osteoclastogenesis via the downregulation of Nfatc1.24 Kim et al. (2019) reported similar outcomes in murine bone marrow macrophage cultures stimulated with MCSF and RANKL. Minocycline or tigecycline (1 / 2.5 / 5 μM) treatment abrogated osteoclast formation through suppression of NFATc1 signaling. The authors speculated that minocycline and tigecycline inhibit osteoclast differentiation by suppressing MMP-9-mediated histone H3 cleavage.25 These reports provide evidence that NFATc1 and MMP-9 may contribute to tetracycline inhibitory actions on osteoclastogenesis.

8. Impact of Tetracyclines on Osteoblastogenesis

The effects of tetracycline antibiotics on osteoblasts (Table 6) have received far less attention than osteoclasts (Table 5). Duewelhenke et al. (2007) found that incubating primary human osteoblasts with tetracycline (60–80 μg/ml) inhibited cell proliferation by 20%.26 Gomes et al. (2007) investigated the effect of doxycycline (1–25 μg/ml) and minocycline (1–50 μg/ml) on the proliferation, differentiation, and function of human bone marrow-derived osteoblastic cells in culture. A 1 μg/ml dose of doxycycline or minocycline increased the proliferation of osteoblastic cells without altering their functional activity. However, higher doses (≥5 μg/ml) inhibited osteoblast function.27 Park et al. (2011) assessed the effects of tetracycline, minocycline, and doxycycline (10 or 100 μM) on MC3T3-E1 murine osteoprecursor cells. Tetracycline at 10 or 100 μM, doxycycline at 100 μM, and minocycline at 100 uM reduced cell viability and mineralization.28 Rathbone et al. (2011) similarly showed that high concentrations of doxycycline and minocycline suppress human osteoblast cell numbers and ALPase activity in vitro.29 Kim et al. (2019) studied minocycline and tigecycline effects on primary murine calvaria derived osteoblasts. Whereas minocycline (2.5 or 5 μM) had no effect, tigecycline (2.5 or 5 μM) attenuated osteoblast function.25 Interestingly, these investigations indicate that tetracycline antibiotics can have divergent dose-dependent effects on osteoblastogenesis.

Table 6:

Tetracycline class antibiotic effects on in vitro osteoblastogenesis

Study Cell Culture System Antibiotic Dose Treatment Duration Outcomes
Duewelhenke et al. (2007)26 Primary human femur derived mesenchymal stem cells stimulated with 100 nM dexamethasone, 50 uM ascorbic acid, & 10 mM β-glycerophosphate Tetracycline 60–80 μg/ml 48 hours
  • ↓ Osteoblast proliferation

Gomes et al. (2007)27 Primary human bone marrow derived osteoblastic cells stimulated with 50 mg/ml ascorbic acid, 10mM β-glycerophosphate, & 10 nM dexamethasone Minocycline or doxycycline Minocycline: 1, 5, 10, 25, 50 μg/ml
Doxycycline: 1, 5, 10, 25 μg/ml
35 days
  • ↑ Osteoblast proliferation induced by 1 & 5 μg/ml doxycycline or 1, 5, & 10 μg/ml minocycline

  • ↓ Osteoblast proliferation induced by 10 & 25 μg/ml doxycycline or 25 & 50 μg/ml minocycline

  • ↓ Osteoblast function induced by 25 μg/ml doxycycline or 50 μg/ml minocycline

Park et al. (2011)28 MC3T3-E1 murine osteoprecursor cells stimulated with 50 μg/mL ascorbic acid & 10 mM β-glycerophosphate Tetracycline, minocycline, or doxycycline 10 or 100 μM 4 days
  • ↓ Osteoblast viability and mineralization induced by 10 & 100 μM tetracycline, 100 μM minocycline, or 100 μM doxycycline

Rathbone et al. (2011)29 Human osteoblasts cultured with 50 μg/ml ascorbic acid, 5 mM glycerophosphate, & 10 nM dexamethasone Minocycline or doxycycline Minocycline: 10, 100, 200, 500 μg/ml
Doxycycline: 10, 100, 200 μg/ml
10 or 14 days
  • ↓ Osteoblast numbers and function induced by 10 – 500 μg/ml minocycline or 10 – 200 μg/ml doxycycline

Kim et al. (2019)25 Primary murine calvaria derived osteoblasts cultured in 50 μg/mL ascorbic acid & 10 mM β-glycerophosphate Minocycline or tigecycline 2.5 or 5 μM 14 or 19 days
  • Minocycline did not alter osteoblast function

  • ↓ Osteoblast function induced by 2.5 & 5 μM tigecycline

9. Future Directions

9.1. Skeletal retention of tetracyclines

Tetracyclines have a high affinity for the bone matrix and are retained within the skeleton for extended periods after discontinuing treatment10,11. This raises the question whether tetracyclines retained in the bone matrix can have prolonged effects on bone modeling and remodeling processes.72,73 Recognizing that long durations of therapy results in cumulative retention in the skeleton,10,11 tetracyclines administered for acne are likely retained in the bone matrix at high concentrations. Roughly 10% of the adult human skeleton is remodeled each year, which includes about 30% of trabecular bone and 3% of cortical bone.147149 Given that tetracyclines are prominently deposited on trabecular surfaces8,9 due to the higher remodeling rate of cancellous bone,147,149 this highlights the potential for tetracycline drugs released from the bone matrix to have lasting effects. Investigations are needed to understand mechanisms by which tetracycline drugs bind the bone matrix, the concentration of drug retained in the skeleton, and how drug / metabolite release from the bone impacts bone modeling / remodeling processes.

9.2. Skeletal maturation and homeostasis

The impact of tetracyclines on bone mass accrual and skeletal maturation during the adolescent and young adult phase is currently unknown. Considering that roughly 40% of peak bone mass is accrued during adolescence and young adulthood,150153 the administration of tetracycline antibiotics during this developmental period could dysregulate skeletal growth and maturation. The effects of tetracycline antibiotics on bone remodeling and homeostasis are currently unclear in the mature adult skeleton. Appreciating that tetracycline drugs are administered for extended durations as a treatment for rosacea in adults, there may be detrimental consequences for bone remodeling and the maintenance of bone mass in the healthy adult skeleton.

9.3. Signaling at bone cells

The mechanisms by which tetracyclines affect bone cells are not well understood. Early investigators studying minocycline-induced changes in the levels of cytosolic Ca2+ in osteoclast cultures speculated that tetracyclines have a partial agonistic effect on the osteoclast Ca2+ receptor.21,22 Minocycline and doxycycline have been reported to inhibit RANKL-induced osteoclastogenesis by blocking downstream MMP-9 enzyme activity, but it is unclear whether this inhibition occurs through the NFATc1 signaling pathway.24,25,146 The signaling mechanisms by which tetracyclines influence osteoblastogenesis have not been investigated. The impact of tetracyclines on osteocytes, which account for 90% of bone cells, also has not been explored. Considering that osteocytes express biologic factors that critically regulate osteoblast and osteoclast activity,154,155 future investigations need to delineate the impact of tetracyclines on osteocyte biology and coupled osteoblast-osteoclast bone remodeling processes.

In vitro investigations have shown that tetracyclines have direct effects on osteoclasts and osteoblasts, but many of these study outcomes were dependent on supraphysiologic drug concentrations (Table 5 and 6). Clinical pharmacokinetic data indicates that long-term administration of doxycycline or minocycline (100 mg every 12 hours) results in steady-state serum concentrations ranging from 1.0 to 3.5 μg/ml.156 Future in vitro bone cell investigations should utilize tetracycline concentrations within the range detected in human serum for study findings to be clinically relevant.

Clinical pharmacokinetic studies have shown that tetracycline drugs are broken down into metabolites, which may have implications for bone cell signaling. There is one known metabolite of tetracycline that has been found in humans, Δ-epitetracycline.52 Whereas there are no known metabolites of doxycycline, six metabolites of minocycline have been detected in humans. The three major minocycline metabolites are 9-Hydroxyminocycline, N7-Demethylminocycline, and N4-Demethylminocycline.52,156158 In order to advance knowledge about the signaling of tetracyclines at bone cells, future studies must begin to investigate how tetracycline drug metabolites impact bone cell physiology.

9.4. Gut microbiota and bone

The link between gut microbiota and bone has recently been recognized. Investigations comparing germ-free mice to conventional mice have shown that immunomodulatory actions imparted by the gut microbiota regulate the development and homeostasis of skeletal tissues159163. Disruption of the gut microbiota by antibiotics has been shown to dysregulate osteoimmune crosstalk in post-pubertal skeletal maturation. A broad-spectrum antibiotic cocktail (vancomycin, imipenem/cilastatin, and neomycin) administered to sex-matched C57BL/6T mice from age 6 to 12 weeks induced phylum shifts in the gut bacteriome and led to suppression of bone mass accrual in the late growing skeleton164. This work indicates that antibiotic-induced changes in the gut microbiota can have indirect effects on the skeleton. Research is needed to determine whether tetracycline drug-induced perturbations in the gut microbiota are linked to alterations in skeletal maturation and homeostasis.

9.5. Third generation tetracycline antibiotic effects on bone

The effects of newer generation tetracycline antibiotics on the skeleton are unclear. Tigecycline is typically administered for infections resistant to earlier generation tetracycline antibiotics.45,46 Several in vitro investigations have begun to evaluate the efficacy of tigecycline for the treatment of osteomyelitis,165167 but the effects of the drug on normal bone physiology is poorly understood. Sarecycline was recently FDA approved for the treatment of moderate-to-severe acne in patients older than nine years of age.41 Considering that sarecycline is administered for 3 to 12 months for the treatment of acne in adolescents and young adults, there could be implications for late skeletal growth and maturation. There are currently no known studies that have evaluated the impact of sarecycline, omadacycline, or eravacycline on bone cells or skeletal metabolism.

10. Conclusion

The mechanisms by which tetracycline drugs bind and are released from the bone matrix is not well understood. The effects of tetracycline, minocycline and doxycycline on bone cell signaling and their impact on physiologic skeletal maturation and homeostasis are unclear. The role of the gut microbiota in tetracycline antibiotic effects on bone metabolism has yet to be investigated.

Newer third generation tetracycline antibiotics (i.e., sarecycline, omadacycline, eravacycline) are being utilized for clinical care, but their effects on bone have not been studied. Future research is needed to elucidate the molecular underpinnings of tetracycline drug actions on bone cells and their impact on skeletal physiology. Tetracyclines’ affinity for and retention in the bone matrix underscores that these drugs may have lasting effects on bone metabolism.

Highlights.

  • Tetracyclines retained in bone may have lasting effects on skeletal metabolism.

  • Tetracyclines’ impact on skeletal maturation and homeostasis is poorly understood.

  • Mechanisms regulating tetracyclines’ actions on bone cells are largely unknown.

  • The gut microbiota’s role in tetracycline antibiotic actions on bone is undefined.

Acknowledgments

The authors thanks Carrie-Anne Warner for graphical artwork instruction and assistance.

Funding Sources

This work was supported by NIH/NIDCR K08DE025337, American Society for Bone and Mineral Research Rising Star Award, NIH/NIDCR T32DE017551, NIH/NIGMS P20GM130457, NIH/NIGMS P20GM121342, NIH/NIDDK P30DK123704, NIH/NIDCR R01DE029637, NIH/NIA R01AG067510.

Abbreviations

MCSF

Macrophage Colony-Stimulating Factor

RANKL

Receptor Activator of Nuclear Factor κβ Ligand

OVX

Ovariectomized

STZ

Streptozotocin

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Nelson ML & Levy SB The history of the tetracyclines. Ann N Y Acad Sci 1241, 17–32, doi: 10.1111/j.1749-6632.2011.06354.x (2011). [DOI] [PubMed] [Google Scholar]
  • 2.Grossman TH Tetracycline Antibiotics and Resistance. Cold Spring Harbor perspectives in medicine 6, a025387, doi: 10.1101/cshperspect.a025387 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Alikhan A, Kurek L & Feldman SR The role of tetracyclines in rosacea. American journal of clinical dermatology 11, 79–87, doi: 10.2165/11530200-000000000-00000 (2010). [DOI] [PubMed] [Google Scholar]
  • 4.Patel DJ & Bhatia N Oral Antibiotics for Acne. American journal of clinical dermatology 22, 193–204, doi: 10.1007/s40257-020-00560-w (2021). [DOI] [PubMed] [Google Scholar]
  • 5.Rusu A & Buta EL The Development of Third-Generation Tetracycline Antibiotics and New Perspectives. Pharmaceutics 13, doi: 10.3390/pharmaceutics13122085 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Golub LM & Lee HM Periodontal therapeutics: Current host-modulation agents and future directions. Periodontology 2000 82, 186–204, doi: 10.1111/prd.12315 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Andre T Studies on the distribution of tritium-labelled dihydrostreptomycin and tetracycline in the body. Acta Radiol Suppl, 1–89 (1956). [PubMed] [Google Scholar]
  • 8.Milch RA, Rall DP & Tobie JE Bone localization of the tetracyclines. Journal of the National Cancer Institute 19, 87–93 (1957). [PubMed] [Google Scholar]
  • 9.Milch RA, Rall DP & Tobie JE Fluorescence of tetracycline antibiotics in bone. The Journal of bone and joint surgery. American volume 40-a, 897–910 (1958). [PubMed] [Google Scholar]
  • 10.Buyske DA, Eisner HJ & Kelly RG Concentration and persistence of tetracycline and chlortetracycline in bone. The Journal of pharmacology and experimental therapeutics 130, 150–156 (1960). [PubMed] [Google Scholar]
  • 11.Myers HM & Jaffe SN TETRACYCLINE BINDING BY SKELETAL TISSUE. Journal of dental research 44, 502–505, doi: 10.1177/00220345650440031001 (1965). [DOI] [PubMed] [Google Scholar]
  • 12.Bevelander G, Nakahara H & Rolle GK Inhibition of skeletal formation in the chick embryo following administration of tetracycline. Nature 184(Suppl 10), 728–729, doi: 10.1038/184728b0 (1959). [DOI] [PubMed] [Google Scholar]
  • 13.Bevelander G, Nakahara H & Rolle GK The effect of tetracycline on the development of the skeletal system of the chick embryo. Developmental biology 2, 298–312, doi: 10.1016/0012-1606(60)90011-7 (1960). [DOI] [PubMed] [Google Scholar]
  • 14.Bevelander G & Cohlan SQ The effect on the rat fetus of transplacentally acquired tetracycline. Biologia neonatorum. Neo-natal studies 4, 365–370, doi: 10.1159/000239846 (1962). [DOI] [PubMed] [Google Scholar]
  • 15.COHLAN SQ, BEVELANDER G & TIAMSIC T Growth Inhibition of Prematures Receiving Tetracycline: A Clinical and Laboratory Investigation of Tetracycline-Induced Bone Fluorescence. American Journal of Diseases of Children 105, 453–461, doi: 10.1001/archpedi.1963.02080040455005 (1963). [DOI] [Google Scholar]
  • 16.Williams S et al. Minocycline prevents the decrease in bone mineral density and trabecular bone in ovariectomized aged rats. Bone 19, 637–644, doi: 10.1016/s8756-3282(96)00302-x (1996). [DOI] [PubMed] [Google Scholar]
  • 17.Porter PJ, Sweeney EA, Golan H & Kass EH Controlled study of the effect of prenatal tetracycline on primary dentition. Antimicrob Agents Chemother (Bethesda) 5, 668–671 (1965). [PubMed] [Google Scholar]
  • 18.Genot MT, Golan HP, Porter PJ & Kass EH Effect of administration of tetracycline in pregnancy on the primary dentition of the offspring. J Oral Med 25, 75–79 (1970). [PubMed] [Google Scholar]
  • 19.Li QN et al. Effects of low doses of hydrochloride tetracycline on bone metabolism and uterus in ovariectomized rats. Acta Pharmacol Sin 24, 599–604 (2003). [PubMed] [Google Scholar]
  • 20.Pytlik M, Folwarczna J & Janiec W Effects of doxycycline on mechanical properties of bones in rats with ovariectomy-induced osteopenia. Calcif Tissue Int 75, 225–230, doi: 10.1007/s00223-004-0097-x (2004). [DOI] [PubMed] [Google Scholar]
  • 21.Bax CM et al. Tetracyclines modulate cytosolic Ca2+ responses in the osteoclast associated with “Ca2+ receptor” activation. Biosci Rep 13, 169–174, doi: 10.1007/bf01149961 (1993). [DOI] [PubMed] [Google Scholar]
  • 22.Zaidi M et al. The effect of tetracyclines on quantitative measures of osteoclast morphology. Biosci Rep 13, 175–182, doi: 10.1007/bf01149962 (1993). [DOI] [PubMed] [Google Scholar]
  • 23.Holmes SG, Still K, Buttle DJ, Bishop NJ & Grabowski PS Chemically modified tetracyclines act through multiple mechanisms directly on osteoclast precursors. Bone 35, 471–478, doi: 10.1016/j.bone.2004.02.028 (2004). [DOI] [PubMed] [Google Scholar]
  • 24.Nagasawa T, Arai M & Togari A Inhibitory effect of minocycline on osteoclastogenesis in mouse bone marrow cells. Arch Oral Biol 56, 924–931, doi: 10.1016/j.archoralbio.2011.02.002 (2011). [DOI] [PubMed] [Google Scholar]
  • 25.Kim Y et al. Tetracycline Analogs Inhibit Osteoclast Differentiation by Suppressing MMP-9-Mediated Histone H3 Cleavage. Int J Mol Sci 20, doi: 10.3390/ijms20164038 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Duewelhenke N, Krut O & Eysel P Influence on mitochondria and cytotoxicity of different antibiotics administered in high concentrations on primary human osteoblasts and cell lines. Antimicrob Agents Chemother 51, 54–63, doi: 10.1128/aac.00729-05 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Gomes PS & Fernandes MH Effect of therapeutic levels of doxycycline and minocycline in the proliferation and differentiation of human bone marrow osteoblastic cells. Arch Oral Biol 52, 251–259, doi: 10.1016/j.archoralbio.2006.10.005 (2007). [DOI] [PubMed] [Google Scholar]
  • 28.Park JB Low dose of doxycyline promotes early differentiation of preosteoblasts by partially regulating the expression of estrogen receptors. J Surg Res 178, 737–742, doi: 10.1016/j.jss.2012.03.072 (2012). [DOI] [PubMed] [Google Scholar]
  • 29.Rathbone CR, Cross JD, Brown KV, Murray CK & Wenke JC Effect of various concentrations of antibiotics on osteogenic cell viability and activity. J Orthop Res 29, 1070–1074, doi: 10.1002/jor.21343 (2011). [DOI] [PubMed] [Google Scholar]
  • 30.Duggar BM Aureomycin; a product of the continuing search for new antibiotics. Ann N Y Acad Sci 51, 177–181, doi: 10.1111/j.1749-6632.1948.tb27262.x (1948). [DOI] [PubMed] [Google Scholar]
  • 31.Nguyen F et al. Tetracycline antibiotics and resistance mechanisms. Biological Chemistry 395, 559–575, doi: 10.1515/hsz-2013-0292 (2014). [DOI] [PubMed] [Google Scholar]
  • 32.Aminov R History of antimicrobial drug discovery: Major classes and health impact. Biochem Pharmacol 133, 4–19, doi: 10.1016/j.bcp.2016.10.001 (2017). [DOI] [PubMed] [Google Scholar]
  • 33.Roberts MC Tetracycline resistance determinants: mechanisms of action, regulation of expression, genetic mobility, and distribution. FEMS Microbiology Reviews 19, 1–24, doi: 10.1111/j.1574-6976.1996.tb00251.x (1996). [DOI] [PubMed] [Google Scholar]
  • 34.Redin GS Antibacterial activity in mice of minocycline, a new tetracycline. Antimicrob Agents Chemother (Bethesda) 6, 371–376 (1966). [PubMed] [Google Scholar]
  • 35.Stephens CR et al. 6-Deoxytetracyclines. IV.1,2 Preparation, C-6 Stereochemistry, and Reactions. Journal of the American Chemical Society 85, 2643–2652, doi: 10.1021/ja00900a027 (1963). [DOI] [Google Scholar]
  • 36.Meijer LA, Ceyssens KG, de Grève BI & de Bruijn W Pharmacokinetics and bioavailability of doxycycline hyclate after oral administration in calves. Vet Q 15, 1–5, doi: 10.1080/01652176.1993.9694358 (1993). [DOI] [PubMed] [Google Scholar]
  • 37.Sum PE & Petersen P Synthesis and structure-activity relationship of novel glycylcycline derivatives leading to the discovery of GAR-936. Bioorg Med Chem Lett 9, 1459–1462, doi: 10.1016/s0960-894x(99)00216-4 (1999). [DOI] [PubMed] [Google Scholar]
  • 38.Livermore DM Tigecycline: what is it, and where should it be used? Journal of Antimicrobial Chemotherapy 56, 611–614, doi: 10.1093/jac/dki291 (2005). [DOI] [PubMed] [Google Scholar]
  • 39.MacGowan AP Tigecycline pharmacokinetic/pharmacodynamic update. Journal of Antimicrobial Chemotherapy 62, i11–i16, doi: 10.1093/jac/dkn242 (2008). [DOI] [PubMed] [Google Scholar]
  • 40.Honeyman L et al. Structure-activity relationship of the aminomethylcyclines and the discovery of omadacycline. Antimicrob Agents Chemother 59, 7044–7053, doi: 10.1128/aac.01536-15 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Andrei S, Droc G & Stefan G FDA approved antibacterial drugs: 2018–2019. Discoveries (Craiova) 7, e102, doi: 10.15190/d.2019.15 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Batool Z, Lomakin IB, Polikanov YS & Bunick CG Sarecycline interferes with tRNA accommodation and tethers mRNA to the 70S ribosome. Proc Natl Acad Sci U S A 117, 20530–20537, doi: 10.1073/pnas.2008671117 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Tan HH Antibacterial therapy for acne: a guide to selection and use of systemic agents. American journal of clinical dermatology 4, 307–314 (2003). [DOI] [PubMed] [Google Scholar]
  • 44.Eichenfield LF et al. Evidence-based recommendations for the diagnosis and treatment of pediatric acne. Pediatrics 131 Suppl 3, S163–186, doi: 10.1542/peds.2013-0490B (2013). [DOI] [PubMed] [Google Scholar]
  • 45.McGovern PC, Wible M, El-Tahtawy A, Biswas P & Meyer RD All-cause mortality imbalance in the tigecycline phase 3 and 4 clinical trials. Int J Antimicrob Agents 41, 463–467, doi: 10.1016/j.ijantimicag.2013.01.020 (2013). [DOI] [PubMed] [Google Scholar]
  • 46.Ahern PP, Faith JJ & Gordon JI Mining the human gut microbiota for effector strains that shape the immune system. Immunity 40, 815–823, doi: 10.1016/j.immuni.2014.05.012 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Danzeisen JL et al. Succession of the turkey gastrointestinal bacterial microbiome related to weight gain. PeerJ 1, e237, doi: 10.7717/peerj.237 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Newman JV, Zhou J, Izmailyan S & Tsai L Randomized, Double-Blind, Placebo-Controlled Studies of the Safety and Pharmacokinetics of Single and Multiple Ascending Doses of Eravacycline. Antimicrob Agents Chemother 62, doi: 10.1128/aac.01174-18 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Tsuda M et al. Intestinal Bifidobacterium association in germ-free T cell receptor transgenic mice down-regulates dietary antigen-specific immune responses of the small intestine but enhances those of the large intestine. Immunobiology 214, 279–289, doi: 10.1016/j.imbio.2008.09.005 (2009). [DOI] [PubMed] [Google Scholar]
  • 50.Zhanel G, Critchley I, Lin LY & Alvandi N Microbiological Profile of Sarecycline, a Novel Targeted Spectrum Tetracycline for the Treatment of Acne Vulgaris. Antimicrob Agents Chemother 63, doi: 10.1128/aac.01297-18 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Shutter MC & Akhondi H in StatPearls (StatPearls Publishing StatPearls Publishing LLC., 2020). [Google Scholar]
  • 52.Agwuh KN & MacGowan A Pharmacokinetics and pharmacodynamics of the tetracyclines including glycylcyclines. The Journal of antimicrobial chemotherapy 58, 256–265, doi: 10.1093/jac/dkl224 (2006). [DOI] [PubMed] [Google Scholar]
  • 53.Zaenglein AL et al. Guidelines of care for the management of acne vulgaris. J Am Acad Dermatol 74, 945–973.e933, doi: 10.1016/j.jaad.2015.12.037 (2016). [DOI] [PubMed] [Google Scholar]
  • 54.Workowski KA, Bolan GA, Centers for Disease, C. & Prevention. Sexually transmitted diseases treatment guidelines, 2015. MMWR Recomm Rep 64, 1–137 (2015). [PMC free article] [PubMed] [Google Scholar]
  • 55.Seas C, DuPont HL, Valdez LM & Gotuzzo E Practical guidelines for the treatment of cholera. Drugs 51, 966–973, doi: 10.2165/00003495-199651060-00005 (1996). [DOI] [PubMed] [Google Scholar]
  • 56.Chey WD, Leontiadis GI, Howden CW & Moss SF ACG Clinical Guideline: Treatment of Helicobacter pylori Infection. Am J Gastroenterol 112, 212–239, doi: 10.1038/ajg.2016.563 (2017). [DOI] [PubMed] [Google Scholar]
  • 57.Smiley CJ et al. Evidence-based clinical practice guideline on the nonsurgical treatment of chronic periodontitis by means of scaling and root planing with or without adjuncts. J Am Dent Assoc 146, 525–535, doi: 10.1016/j.adaj.2015.01.026 (2015). [DOI] [PubMed] [Google Scholar]
  • 58.Brogden RN, Speight TM & Avery GS Minocycline: A review of its antibacterial and pharmacokinetic properties and therapeutic use. Drugs 9, 251–291, doi: 10.2165/00003495-197509040-00005 (1975). [DOI] [PubMed] [Google Scholar]
  • 59.Shankar C, Nabarro LEB, Anandan S & Veeraraghavan B Minocycline and Tigecycline: What Is Their Role in the Treatment of Carbapenem-Resistant Gram-Negative Organisms? Microb Drug Resist 23, 437–446, doi: 10.1089/mdr.2016.0043 (2017). [DOI] [PubMed] [Google Scholar]
  • 60.Newton PN et al. Pharmacokinetics of oral doxycycline during combination treatment of severe falciparum malaria. Antimicrob Agents Chemother 49, 1622–1625, doi: 10.1128/aac.49.4.1622-1625.2005 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Hendricks KA et al. Centers for disease control and prevention expert panel meetings on prevention and treatment of anthrax in adults. Emerg Infect Dis 20, doi: 10.3201/eid2002.130687 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Lago K et al. Impact of Doxycycline as Malaria Prophylaxis on Risk of Influenza-Like Illness among International Travelers. Am J Trop Med Hyg 102, 821–826, doi: 10.4269/ajtmh.19-0648 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Meagher AK, Ambrose PG, Grasela TH & Ellis-Grosse EJ Pharmacokinetic/pharmacodynamic profile for tigecycline-a new glycylcycline antimicrobial agent. Diagn Microbiol Infect Dis 52, 165–171, doi: 10.1016/j.diagmicrobio.2005.05.006 (2005). [DOI] [PubMed] [Google Scholar]
  • 64.Ellis-Grosse EJ, Babinchak T, Dartois N, Rose G & Loh E The efficacy and safety of tigecycline in the treatment of skin and skin-structure infections: results of 2 double-blind phase 3 comparison studies with vancomycin-aztreonam. Clin Infect Dis 41 Suppl 5, S341–353, doi: 10.1086/431675 (2005). [DOI] [PubMed] [Google Scholar]
  • 65.Babinchak T, Ellis-Grosse E, Dartois N, Rose GM & Loh E The efficacy and safety of tigecycline for the treatment of complicated intra-abdominal infections: analysis of pooled clinical trial data. Clin Infect Dis 41 Suppl 5, S354–367, doi: 10.1086/431676 (2005). [DOI] [PubMed] [Google Scholar]
  • 66.Information., N. C. f. B. < https://pubchem.ncbi.nlm.nih.gov/compound/Sarecycline.> (
  • 67.in UpToDate (Wolters Kluwer, 2022). [Google Scholar]
  • 68.James WD Clinical practice. Acne. N Engl J Med 352, 1463–1472, doi: 10.1056/NEJMcp033487 (2005). [DOI] [PubMed] [Google Scholar]
  • 69.Leyden JJ, Del Rosso JQ & Webster GF Clinical considerations in the treatment of acne vulgaris and other inflammatory skin disorders: focus on antibiotic resistance. Cutis 79, 9–25 (2007). [PubMed] [Google Scholar]
  • 70.Bickers DR et al. The burden of skin diseases: 2004 a joint project of the American Academy of Dermatology Association and the Society for Investigative Dermatology. Journal of the American Academy of Dermatology 55, 490–500, doi: 10.1016/j.jaad.2006.05.048 (2006). [DOI] [PubMed] [Google Scholar]
  • 71.Bhate K & Williams HC Epidemiology of acne vulgaris. Br J Dermatol 168, 474–485, doi: 10.1111/bjd.12149 (2013). [DOI] [PubMed] [Google Scholar]
  • 72.Lee YH, Liu G, Thiboutot DM, Leslie DL & Kirby JS A retrospective analysis of the duration of oral antibiotic therapy for the treatment of acne among adolescents: investigating practice gaps and potential cost-savings. Journal of the American Academy of Dermatology 71, 70–76, doi: 10.1016/j.jaad.2014.02.031 (2014). [DOI] [PubMed] [Google Scholar]
  • 73.Nagler AR, Milam EC & Orlow SJ The use of oral antibiotics before isotretinoin therapy in patients with acne. Journal of the American Academy of Dermatology 74, 273–279, doi: 10.1016/j.jaad.2015.09.046 (2016). [DOI] [PubMed] [Google Scholar]
  • 74.Del Rosso JQ et al. Status Report from the Scientific Panel on Antibiotic Use in Dermatology of the American Acne and Rosacea Society: Part 1: Antibiotic Prescribing Patterns, Sources of Antibiotic Exposure, Antibiotic Consumption and Emergence of Antibiotic Resistance, Impact of Alterations in Antibiotic Prescribing, and Clinical Sequelae of Antibiotic Use. J Clin Aesthet Dermatol 9, 18–24 (2016). [PMC free article] [PubMed] [Google Scholar]
  • 75.Dreno B Topical antibacterial therapy for acne vulgaris. Drugs 64, 2389–2397, doi: 10.2165/00003495-200464210-00002 (2004). [DOI] [PubMed] [Google Scholar]
  • 76.Krakowski AC, Stendardo S & Eichenfield LF Practical considerations in acne treatment and the clinical impact of topical combination therapy. Pediatr Dermatol 25 Suppl 1, 1–14, doi: 10.1111/j.1525-1470.2008.00667.x (2008). [DOI] [PubMed] [Google Scholar]
  • 77.Esterly NB, Koransky JS, Furey NL & Trevisan M Neutrophil chemotaxis in patients with acne receiving oral tetracycline therapy. Arch Dermatol 120, 1308–1313 (1984). [PubMed] [Google Scholar]
  • 78.Moore A et al. Once-Daily Oral Sarecycline 1.5 mg/kg/day Is Effective for Moderate to Severe Acne Vulgaris: Results from Two Identically Designed, Phase 3, Randomized, Double-Blind Clinical Trials. J Drugs Dermatol 17, 987–996 (2018). [PubMed] [Google Scholar]
  • 79.Pariser DM et al. Safety and Tolerability of Sarecycline for the Treatment of Acne Vulgaris: Results from a Phase III, Multicenter, Open-Label Study and a Phase I Phototoxicity Study. J Clin Aesthet Dermatol 12, E53–e62 (2019). [PMC free article] [PubMed] [Google Scholar]
  • 80.Deeks ED Sarecycline: First Global Approval. Drugs 79, 325–329, doi: 10.1007/s40265-019-1053-4 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Valente Duarte de Sousa, I. C. An overview of sarecycline for the treatment of moderate-to-severe acne vulgaris. Expert Opin Pharmacother, 1–10, doi: 10.1080/14656566.2020.1813279 (2020). [DOI] [PubMed] [Google Scholar]
  • 82.Leyden JJ Antibiotic resistance in the topical treatment of acne vulgaris. Cutis 73, 6–10 (2004). [PubMed] [Google Scholar]
  • 83.Gollnick H et al. Management of acne: a report from a Global Alliance to Improve Outcomes in Acne. Journal of the American Academy of Dermatology 49, S1–37, doi: 10.1067/mjd.2003.618 (2003). [DOI] [PubMed] [Google Scholar]
  • 84.Ozolins M et al. Comparison of five antimicrobial regimens for treatment of mild to moderate inflammatory facial acne vulgaris in the community: randomised controlled trial. Lancet 364, 2188–2195, doi: 10.1016/s0140-6736(04)17591-0 (2004). [DOI] [PubMed] [Google Scholar]
  • 85.Graber EM (UpToDate Inc, Waltham, MA, 2021).
  • 86.in LiverTox: Clinical and Research Information on Drug-Induced Liver Injury (National Institute of Diabetes and Digestive and Kidney Diseases, 2022). [Google Scholar]
  • 87.Mays RM, Gordon RA, Wilson JM & Silapunt S New antibiotic therapies for acne and rosacea. Dermatol Ther 25, 23–37, doi: 10.1111/j.1529-8019.2012.01497.x (2012). [DOI] [PubMed] [Google Scholar]
  • 88.Greenstein G The role of Periostat in the management of adult periodontitis: a critical assessment. Compend Contin Educ Dent 20, 664–668, 670, 672 passim (1999). [PubMed] [Google Scholar]
  • 89.Preshaw PM et al. Subantimicrobial dose doxycycline as adjunctive treatment for periodontitis. A review. Journal of clinical periodontology 31, 697–707, doi: 10.1111/j.1600-051X.2004.00558.x (2004). [DOI] [PubMed] [Google Scholar]
  • 90.Caton J & Ryan ME Clinical studies on the management of periodontal diseases utilizing subantimicrobial dose doxycycline (SDD). Pharmacological research : the official journal of the Italian Pharmacological Society 63, 114–120, doi: 10.1016/j.phrs.2010.12.003 (2011). [DOI] [PubMed] [Google Scholar]
  • 91.van Zuuren EJ Rosacea. N Engl J Med 377, 1754–1764, doi: 10.1056/NEJMcp1506630 (2017). [DOI] [PubMed] [Google Scholar]
  • 92.Berman B & Zell D Subantimicrobial dose doxycycline: a unique treatment for rosacea. Cutis 75, 19–24 (2005). [PubMed] [Google Scholar]
  • 93.Del Rosso JQ Recently approved systemic therapies for acne vulgaris and rosacea. Cutis 80, 113–120 (2007). [PubMed] [Google Scholar]
  • 94.Layton A & Thiboutot D Emerging therapies in rosacea. J Am Acad Dermatol 69, S57–65, doi: 10.1016/j.jaad.2013.04.041 (2013). [DOI] [PubMed] [Google Scholar]
  • 95.Kelly RG & Buyske DA Metabolism of tetracycline in the rat and the dog. The Journal of pharmacology and experimental therapeutics 130, 144–149 (1960). [PubMed] [Google Scholar]
  • 96.Frost HM & Villaneuva AR Tetracycline staining of newly forming bone and mineralizing cartilage in vivo. Stain technology 35, 135–138, doi: 10.3109/10520296009114729 (1960). [DOI] [PubMed] [Google Scholar]
  • 97.Frost HM Human osteoblastic activity. II. Measurement of the biological half-life of bones with the aid of tetracyclines. Henry Ford Hospital medical bulletin 9, 87–96 (1961). [PubMed] [Google Scholar]
  • 98.Frost HM Mean formation time of human osteons. Canadian journal of biochemistry and physiology 41, 1307–1310 (1963). [PubMed] [Google Scholar]
  • 99.Frost HM, Roth H, Villanueva AR & Stanisavljevic S Experimental multiband tetracycline measurement of lamellar osteoblastic activity. Henry Ford Hospital medical bulletin 9, 312–329 (1961). [PubMed] [Google Scholar]
  • 100.Frost HM Measurement of human bone formation by means of tetracycline labelling. Canadian journal of biochemistry and physiology 41, 31–42 (1963). [PubMed] [Google Scholar]
  • 101.Parfitt AM et al. Bone histomorphometry: standardization of nomenclature, symbols, and units. Report of the ASBMR Histomorphometry Nomenclature Committee. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research 2, 595–610, doi: 10.1002/jbmr.5650020617 (1987). [DOI] [PubMed] [Google Scholar]
  • 102.Dempster DW et al. Standardized nomenclature, symbols, and units for bone histomorphometry: a 2012 update of the report of the ASBMR Histomorphometry Nomenclature Committee. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research 28, 2–17, doi: 10.1002/jbmr.1805 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Allen Matthew R., D. B. B. in Basic and Applied Bone Biology (ed Allen Matthew R. Burr David B.) 131–148 (2014). [Google Scholar]
  • 104.Wolfe ID & Reichmister J Minocycline hyperpigmentation: skin, tooth, nail, and bone involvement. Cutis 33, 457–458 (1984). [PubMed] [Google Scholar]
  • 105.Fendrich P & Brooke RI An unusual case of oral pigmentation. Oral surgery, oral medicine, and oral pathology 58, 288–289, doi: 10.1016/0030-4220(84)90056-2 (1984). [DOI] [PubMed] [Google Scholar]
  • 106.Middleton SD, Anakwe RE & McKinley JC Black bone disease of the foot. Minocycline related pigmentation. Foot Ankle Surg 17, e34–36, doi: 10.1016/j.fas.2011.01.011 (2011). [DOI] [PubMed] [Google Scholar]
  • 107.Reed DN, Gregg FO & Corpe RS Minocycline-induced black bone disease encountered during total knee arthroplasty. Orthopedics 35, e737–739, doi: 10.3928/01477447-20120426-30 (2012). [DOI] [PubMed] [Google Scholar]
  • 108.Kerbleski GJ, Hampton TT & Cornejo A Black bone disease of the foot: a case study and review of literature demonstrating a correlation of long-term minocycline therapy and bone hyperpigmentation. The Journal of foot and ankle surgery : official publication of the American College of Foot and Ankle Surgeons 52, 239–241, doi: 10.1053/j.jfas.2012.10.018 (2013). [DOI] [PubMed] [Google Scholar]
  • 109.Odell EW, Hodgson RP & Haskell R Oral presentation of minocycline-induced black bone disease. Oral surgery, oral medicine, oral pathology, oral radiology, and endodontics 79, 459–461, doi: 10.1016/s1079-2104(05)80128-3 (1995). [DOI] [PubMed] [Google Scholar]
  • 110.Salman RA, Salman DG, Glickman RS, Super S & Salman L Minocycline induced pigmentation of the oral cavity. J Oral Med 40, 154–157 (1985). [PubMed] [Google Scholar]
  • 111.Cale AE, Freedman PD & Lumerman H Pigmentation of the jawbones and teeth secondary to minocycline hydrochloride therapy. Journal of periodontology 59, 112–114, doi: 10.1902/jop.1988.59.2.112 (1988). [DOI] [PubMed] [Google Scholar]
  • 112.Westbury LW & Najera A Minocycline-induced intraoral pharmacogenic pigmentation: case reports and review of the literature. J Periodontol 68, 84–91, doi: 10.1902/jop.1997.68.1.84 (1997). [DOI] [PubMed] [Google Scholar]
  • 113.Ayangco L & Sheridan PJ Minocycline-induced staining of torus palatinus and alveolar bone. J Periodontol 74, 669–671, doi: 10.1902/jop.2003.74.5.669 (2003). [DOI] [PubMed] [Google Scholar]
  • 114.Damm DD & Fantasia JE Blue palate and alveolar ridge. General dentistry 52, 554, 556 (2004). [PubMed] [Google Scholar]
  • 115.Treister NS, Magalnick D & Woo SB Oral mucosal pigmentation secondary to minocycline therapy: report of two cases and a review of the literature. Oral surgery, oral medicine, oral pathology, oral radiology, and endodontics 97, 718–725, doi: 10.1016/j.tripleo.2003.11.006 (2004). [DOI] [PubMed] [Google Scholar]
  • 116.Rumbak MJ, Pitcock JA, Palmieri GM & Robertson JT Black bones following long-term minocycline treatment. Archives of pathology & laboratory medicine 115, 939–941 (1991). [PubMed] [Google Scholar]
  • 117.McCleskey PE & Littleton KH Minocycline-induced blue-green discoloration of bone. A case report. The Journal of bone and joint surgery. American volume 86, 146–148, doi: 10.2106/00004623-200401000-00023 (2004). [DOI] [PubMed] [Google Scholar]
  • 118.Pandit S & Hadden W Black pigmentation of bone due to long-term minocycline use. Surgeon 2, 236–237, doi: 10.1016/s1479-666x(04)80008-8 (2004). [DOI] [PubMed] [Google Scholar]
  • 119.Hepburn MJ, Dooley DP & Hayda RA Minocycline-induced black bone disease. Orthopedics 28, 501–502 (2005). [DOI] [PubMed] [Google Scholar]
  • 120.Somayazula R & Rogers GF Metacarpal darkening associated with minocycline therapy. The Journal of hand surgery, European volume 35, 760–761, doi: 10.1177/1753193410378953 (2010). [DOI] [PubMed] [Google Scholar]
  • 121.Yang S et al. Minocycline-induced periarticular black bones in inflamed joints which underwent arthroplastic reconstruction. Clin Orthop Surg 4, 181–187, doi: 10.4055/cios.2012.4.3.181 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Kelly RG & Kanegis LA Metabolism and tissue distribution of radioisotopically labelled minocycline. Toxicology and applied pharmacology 11, 171–183, doi: 10.1016/0041-008x(67)90036-1 (1967). [DOI] [PubMed] [Google Scholar]
  • 123.Hughes WH, Lee WR & Flood DJ A comparative study of the actions of six tetracyclines on the development of the chick embryo. Br J Pharmacol Chemother 25, 317–323, doi: 10.1111/j.1476-5381.1965.tb02052.x (1965). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Johnson RH & Mitchell DF The effects of tetracyclines on teeth and bones. J Dent Res 45, 86–93, doi: 10.1177/00220345660450013601 (1966). [DOI] [PubMed] [Google Scholar]
  • 125.Charles D Placental transmission of antibiotics. J Obstet Gynaecol Br Emp 61, 750–757, doi: 10.1111/j.1471-0528.1954.tb07720.x (1954). [DOI] [PubMed] [Google Scholar]
  • 126.Gibbons RJ & Reichelderfer TE Transplacental transmission of demethylchlortetracycline and toxicity studies in premature and full term, newly born infants. Antibiotic Med Clin Ther (New York) 7, 618–622 (1960). [PubMed] [Google Scholar]
  • 127.Pugashetti R & Shinkai K Treatment of acne vulgaris in pregnant patients. Dermatol Ther 26, 302–311, doi: 10.1111/dth.12077 (2013). [DOI] [PubMed] [Google Scholar]
  • 128.Johnson RH THE TETRACYCLINES: A REVIEW OF THE LITERATURE--1948 THROUGH 1963. J Oral Ther Pharmacol 1, 190–217 (1964). [PubMed] [Google Scholar]
  • 129.Wormser GP et al. The clinical assessment, treatment, and prevention of lyme disease, human granulocytic anaplasmosis, and babesiosis: clinical practice guidelines by the Infectious Diseases Society of America. Clin Infect Dis 43, 1089–1134, doi: 10.1086/508667 (2006). [DOI] [PubMed] [Google Scholar]
  • 130.Chung AM, Reed MD & Blumer JL Antibiotics and breast-feeding: a critical review of the literature. Paediatr Drugs 4, 817–837, doi: 10.2165/00128072-200204120-00006 (2002). [DOI] [PubMed] [Google Scholar]
  • 131.Wallman IS & Hilton HB Teeth pigmented by tetracycline. Lancet 1, 827–829, doi: 10.1016/s0140-6736(62)91840-8 (1962). [DOI] [PubMed] [Google Scholar]
  • 132.Tredwin CJ, Scully C & Bagan-Sebastian JV Drug-induced disorders of teeth. Journal of dental research 84, 596–602, doi: 10.1177/154405910508400703 (2005). [DOI] [PubMed] [Google Scholar]
  • 133.Sánchez AR, Rogers RS 3rd & Sheridan PJ Tetracycline and other tetracycline-derivative staining of the teeth and oral cavity. International journal of dermatology 43, 709–715, doi: 10.1111/j.1365-4632.2004.02108.x (2004). [DOI] [PubMed] [Google Scholar]
  • 134.de Figueiredo FAT et al. Doxycycline reduces osteopenia in female rats. Sci Rep 9, 15316, doi: 10.1038/s41598-019-51702-y (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Golub LM et al. Tetracycline administration prevents diabetes-induced osteopenia in the rat: initial observations. Research communications in chemical pathology and pharmacology 68, 27–40 (1990). [PubMed] [Google Scholar]
  • 136.Fowlkes JL et al. Effects of long-term doxycycline on bone quality and strength in diabetic male DBA/2J mice. Bone Rep 1, 16–19, doi: 10.1016/j.bonr.2014.10.001 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Bain S et al. Tetracycline prevents cancellous bone loss and maintains near-normal rates of bone formation in streptozotocin diabetic rats. Bone 21, 147–153, doi: 10.1016/s8756-3282(97)00104-x (1997). [DOI] [PubMed] [Google Scholar]
  • 138.Sasaki T, Kaneko H, Ramamurthy NS & Golub LM Tetracycline administration restores osteoblast structure and function during experimental diabetes. Anat Rec 231, 25–34, doi: 10.1002/ar.1092310105 (1991). [DOI] [PubMed] [Google Scholar]
  • 139.Sasaki T, Ramamurthy NS & Golub LM Tetracycline administration increases collagen synthesis in osteoblasts of streptozotocin-induced diabetic rats: a quantitative autoradiographic study. Calcif Tissue Int 50, 411–419, doi: 10.1007/bf00296771 (1992). [DOI] [PubMed] [Google Scholar]
  • 140.Kaneko H, Sasaki T, Ramamurthy NS & Golub LM Tetracycline administration normalizes the structure and acid phosphatase activity of osteoclasts in streptozotocin-induced diabetic rats. The Anatomical record 227, 427–436, doi: 10.1002/ar.1092270406 (1990). [DOI] [PubMed] [Google Scholar]
  • 141.Donahue HJ, Iijima K, Goligorsky MS, Rubin CT & Rifkin BR Regulation of cytoplasmic calcium concentration in tetracycline-treated osteoclasts. J Bone Miner Res 7, 1313–1318, doi: 10.1002/jbmr.5650071111 (1992). [DOI] [PubMed] [Google Scholar]
  • 142.Gomes BC, Golub LM & Ramamurthy NS Tetracyclines inhibit parathyroid hormone-induced bone resorption in organ culture. Experientia 40, 1273–1275, doi: 10.1007/bf01946671 (1984). [DOI] [PubMed] [Google Scholar]
  • 143.Zhou X, Zhang P, Zhang C, An B & Zhu Z Tetracyclines inhibit rat osteoclast formation and activity in vitro and affect bone turnover in young rats in vivo. Calcif Tissue Int 86, 163–171, doi: 10.1007/s00223-009-9328-5 (2010). [DOI] [PubMed] [Google Scholar]
  • 144.Chowdhury MH, Moak SA, Rifkin BR & Greenwald RA Effect of tetracyclines which have metalloproteinase inhibitory capacity on basal and heparin-stimulated bone resorption by chick osteoclasts. Agents Actions 40, 124–128, doi: 10.1007/bf01976761 (1993). [DOI] [PubMed] [Google Scholar]
  • 145.Bettany JT, Peet NM, Wolowacz RG, Skerry TM & Grabowski PS Tetracyclines induce apoptosis in osteoclasts. Bone 27, 75–80, doi: 10.1016/s8756-3282(00)00297-0 (2000). [DOI] [PubMed] [Google Scholar]
  • 146.Franco GC et al. Inhibition of matrix metalloproteinase-9 activity by doxycycline ameliorates RANK ligand-induced osteoclast differentiation in vitro and in vivo. Exp Cell Res 317, 1454–1464, doi: 10.1016/j.yexcr.2011.03.014 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Parfitt AM Osteonal and hemi-osteonal remodeling: the spatial and temporal framework for signal traffic in adult human bone. Journal of cellular biochemistry 55, 273–286, doi: 10.1002/jcb.240550303 (1994). [DOI] [PubMed] [Google Scholar]
  • 148.Sims NA & Martin TJ Coupling the activities of bone formation and resorption: a multitude of signals within the basic multicellular unit. BoneKEy reports 3, 481, doi: 10.1038/bonekey.2013.215 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Manolagas SC Birth and death of bone cells: basic regulatory mechanisms and implications for the pathogenesis and treatment of osteoporosis. Endocrine reviews 21, 115–137, doi: 10.1210/edrv.21.2.0395 (2000). [DOI] [PubMed] [Google Scholar]
  • 150.McCormack SE et al. Association Between Linear Growth and Bone Accrual in a Diverse Cohort of Children and Adolescents. JAMA pediatrics 171, e171769, doi: 10.1001/jamapediatrics.2017.1769 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Weaver CM et al. The National Osteoporosis Foundation’s position statement on peak bone mass development and lifestyle factors: a systematic review and implementation recommendations. Osteoporosis international : a journal established as result of cooperation between the European Foundation for Osteoporosis and the National Osteoporosis Foundation of the USA 27, 1281–1386, doi: 10.1007/s00198-015-3440-3 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Baxter-Jones AD, Faulkner RA, Forwood MR, Mirwald RL & Bailey DA Bone mineral accrual from 8 to 30 years of age: an estimation of peak bone mass. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research 26, 1729–1739, doi: 10.1002/jbmr.412 (2011). [DOI] [PubMed] [Google Scholar]
  • 153.Bonjour JP, Theintz G, Buchs B, Slosman D & Rizzoli R Critical years and stages of puberty for spinal and femoral bone mass accumulation during adolescence. The Journal of clinical endocrinology and metabolism 73, 555–563, doi: 10.1210/jcem-73-3-555 (1991). [DOI] [PubMed] [Google Scholar]
  • 154.Bonewald LF in Primer on the Metabolic Bone Diseases and Disorders of Mineral Metabolism 38–45 (John Wiley & Sons, Inc. : Hoboken, NJ, USA, 2018). [Google Scholar]
  • 155.Florencio-Silva R, Sasso GR, Sasso-Cerri E, Simoes MJ & Cerri PS Biology of Bone Tissue: Structure, Function, and Factors That Influence Bone Cells. BioMed research international 2015, 421746, doi: 10.1155/2015/421746 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Saivin S & Houin G Clinical pharmacokinetics of doxycycline and minocycline. Clinical pharmacokinetics 15, 355–366, doi: 10.2165/00003088-198815060-00001 (1988). [DOI] [PubMed] [Google Scholar]
  • 157.Nelis HJ & De Leenheer AP Metabolism of minocycline in humans. Drug metabolism and disposition: the biological fate of chemicals 10, 142–146 (1982). [PubMed] [Google Scholar]
  • 158.Böcker RH, Peter R, Machbert G & Bauer W Identification and determination of the two principal metabolites of minocycline in humans. Journal of chromatography 568, 363–374, doi: 10.1016/0378-4347(91)80174-b (1991). [DOI] [PubMed] [Google Scholar]
  • 159.Sjogren K et al. The gut microbiota regulates bone mass in mice. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research 27, 1357–1367, doi: 10.1002/jbmr.1588 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Schwarzer M et al. Lactobacillus plantarum strain maintains growth of infant mice during chronic undernutrition. Science (New York, N.Y.) 351, 854–857, doi: 10.1126/science.aad8588 (2016). [DOI] [PubMed] [Google Scholar]
  • 161.Yan J et al. Gut microbiota induce IGF-1 and promote bone formation and growth. Proceedings of the National Academy of Sciences of the United States of America 113, E7554–e7563, doi: 10.1073/pnas.1607235113 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Novince CM et al. Commensal Gut Microbiota Immunomodulatory Actions in Bone Marrow and Liver have Catabolic Effects on Skeletal Homeostasis in Health. Sci Rep 7, 5747, doi: 10.1038/s41598-017-06126-x (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Li JY et al. Sex steroid deficiency-associated bone loss is microbiota dependent and prevented by probiotics. The Journal of clinical investigation 126, 2049–2063, doi: 10.1172/jci86062 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Hathaway-Schrader JD et al. Antibiotic Perturbation of Gut Microbiota Dysregulates Osteoimmune Cross Talk in Postpubertal Skeletal Development. The American Journal of Pathology 189, 370–390, doi: 10.1016/j.ajpath.2018.10.017 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Kreis CA et al. Therapy of intracellular Staphylococcus aureus by tigecyclin. BMC infectious diseases 13, 267, doi: 10.1186/1471-2334-13-267 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Ignjatović NL, Ninkov P, Sabetrasekh R, Lyngstadaas SP & Uskoković DP In vitro evaluation of a multifunctional nano drug delivery system based on tigecycline-loaded calcium-phosphate/ poly-DL-lactide-co-glycolide. Bio-medical materials and engineering 24, 1647–1658, doi: 10.3233/bme-140978 (2014). [DOI] [PubMed] [Google Scholar]
  • 167.Valour F et al. Antimicrobial activity against intraosteoblastic Staphylococcus aureus. Antimicrobial agents and chemotherapy 59, 2029–2036, doi: 10.1128/aac.04359-14 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES