Content » Vol 97, Issue 2

Special Report

Apoptosis May Explain the Pharmacological Mode of Action and Adverse Effects of Isotretinoin, Including Teratogenicity

Bodo C. Melnik

Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, Osnabrück, Germany

ABSTRACT

Isotretinoin (13-cis retinoic acid) is the most effective sebum-suppressive drug for the treatment of severe acne. Its effect depends on sebocyte apoptosis, which results from isotretinoin-induced expression of the apoptotic protein tumour necrosis factor-related apoptosis-inducing ligand, insulin-like growth factor-binding protein-3 and neutrophil gelatinase-associated lipocalin. This review proposes that the pharmacological mode of action of isotretinoin in the treatment of severe acne, acute promyelocytic leukaemia, and neuroblastoma results from apoptosis. Furthermore, apoptosis may be the underlying and unifying mechanism of the adverse effects of isotretinoin on neural crest cells (teratogenicity), hippocampal neurones (depression), epidermal keratinocytes and mucosa cells (mucocutaneous side-effects), hair follicle cells (telogen effluvium), intestinal epithelial cells (inflammatory bowel disease), skeletal muscle cells (myalgia and release of creatine kinase), and hepatocytes (release of transaminases and very low-density lipoproteins). Genetic variants of components of the apoptotic signalling cascade, such as RARA polymorphisms, might explain variations in the magnitude of isotretinoin-induced apoptotic signalling and apparently identify subgroups of patients who experience either stronger adverse effects with isotretinoin therapy or resistance to treatment.

Key words: adverse effects; apoptosis; FoxOs; isotretinoin; TRAIL.

Accepted Sep 19, 2016; Epub ahead of print Sep 27, 2016

Acta Derm Venereol 2017; 97: XX–XX

Corr: Bodo C. Melnik, Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, Sedanstrasse 115, DE-49090 Osnabück, Germany. E-mail: melnik@t-online.de

INTRODUCTION

Oral isotretinoin (13-cis retinoic acid) is the most effective drug in the treatment of severe nodular recalcitrant acne (1). Isotretinoin was introduced to the market by Hoffman-La Roche in 1982. However, treatment with isotretinoin is associated with adverse effects, the most serious of which is teratogenicity (2, 3). Further manageable adverse effects are mucocutaneous side-effects, increases in transaminases, and hypertriglyceridaemia. Rare adverse effects are depression and inflammatory bowel disease. The aim of this medical hypothesis is to show that the underlying common mechanism that explains the mode of action of isotretinoin and all its adverse effects is apoptosis. The desired apoptotic effect of isotretinoin in the treatment of acne is discussed first, followed by its unwanted adverse effects, including teratogenicity.

SEBOCYTE APOPTOSIS

The major pharmacological action of isotretinoin in the treatment of severe acne is sebum suppression (1). Excessive production of sebum and qualitative proinflammatory changes in sebum composition (“acne sebum”) are thought to play a major role in the pathogenesis of acne (4–6). Of all known anti-acne drugs, isotretinoin exhibits the strongest sebum-suppressive effect (7). This sebum-suppressive activity is primarily based on sebocyte apoptosis. Nelson et al. (8) demonstrated that isotretinoin induces apoptosis and cell cycle arrest in human SEB-1 sebocytes. In 2 further studies, Nelson et al. (9) showed that the apoptotic protein tumour necrosis factor-related apoptosis inducing ligand (TRAIL) and neutrophil gelatinase-associated lipocalin (NGAL) contribute to the apoptotic effect of isotretinoin in human sebaceous gland cells (10). Kelhälä et al. (11) recently confirmed increased expression of TRAIL and NGAL in the skin of patients with acne during treatment with isotretinoin. TRAIL (Apo-2L), which induces apoptosis in a number of tumour cell lines, is relatively non-toxic to normal cells. Consistent with its lack of toxicity, TRAIL is constitutively expressed in many human tissues (12). At the promoter level, TRAIL expression is induced by nuclear FoxO transcription factors, especially FoxO3a (13, 14). In patients with acne, FoxO activity is suppressed due to enhanced growth factor signalling (15–17). Insulin-like growth factor-1 (IGF-1) and insulin, which both increase during puberty and under the influence of Western diet (hyperglycaemic carbohydrates and milk) (18, 19), stimulate the kinase Akt, resulting in FoxO phosphorylation, which leads to nuclear FoxO inactivation through cytoplasmic sequestration via 14-3-3 binding (20, 21). FoxOs promote cell growth inhibitory and/or apoptosis signalling, either by stimulating expression of death receptor ligands, such as TRAIL and Fas ligand (FASL), or by inducing the expression of multiple pro-apoptotic members of the Bcl2 family of mitochondria-targeting proteins, or by enhancing levels of cyclin-dependent kinase inhibitors, such as p21 and p27 (13), as demonstrated in isotretinoin-treated SEB-1 sebocytes (8). In sebocytes, the prodrug isotretinoin (13-cis retinoic acid) is isomerized to all-trans retinoic acid (ATRA) (Fig. 1) (22). ATRA upregulates the expression of FoxO3a and TRAIL (23–25). FoxO3a is a key transcription factor of apoptosis (25). In fact, FoxO3a DNA-binding elements exist in the TRAIL promoter, indicating that TRAIL is a direct target of ATRA-induced FoxO3a (14). TRAIL has been shown to be upregulated in human sebaceous gland cells during treatment with isotretinoin (9–11). Thus, it is reasonable to assume that isotretinoin increases FoxO/TRAIL signalling in sebaceous glands of patients with acne, resulting in sebocyte apoptosis. The resulting suppression of sebum, “Kligman’s oil of the acne flame”, is the major anti-acne action of oral isotretinoin therapy.


Fig. 1. Hypothesis of isotretinoin-induced apoptotic signalling explaining the pharmacological and adverse effects of isotretinoin. After isomerization of isotretinoin to all-trans retinoic acid (ATRA) and ATRA binding to retinoic acid receptor (RAR) the transcription factor FoxO3a is upregulated. At the promoter level FoxO3a induces the expression of tumour necrosis factor-related apoptosis inducing ligand (TRAIL) and FoxO1. TRAIL activates the caspase cascade, resulting in apoptosis. FoxO1 mediates cell cycle arrest via upregulation of the cell cycle inhibitors p21 and p27.

FoxOs also play a critical role in the expression of the pro-apoptotic IGF-binding protein-3 (IGFBP3) (26), which acts as a nuclear transcription factor (27). Treatment with isotretinoin significantly increased IGFBP3 in sebocytes (10). Nuclear IGFBP3 interacts with retinoid X receptor-α (RXRα) and retinoic acid receptor-α (RARα) and thereby interferes with the formation of RXR:RAR heterodimers (28). The RXR-IGFBP3 interaction modulates the transcriptional activity of RXRα and is essential for mediating the effects of IGFBP3 on apoptosis (29). FoxO-dependent induction of nuclear IGFBP3 with consecutive activation of RARα may enhance the expression of pro-apoptotic NGAL (30), whose expression at the promoter level is regulated via RARα and RXRα (10). Thus, there is substantial evidence that isotretinoin increases the expression of the pro-apoptotic FoxO proteins, TRAIL, IGFBP3 and NGAL.

There is good reason to assume that pro-apoptotic isotretinoin signalling balances increased Akt-mediated pro-survival signalling in acne skin (31). Increased IGF-1-driven Akt signalling maintains high expression of the anti-apoptotic protein survivin (32), which is upregulated in the serum and skin of patients with acne (133, added in proof). Notably, it has been shown that FoxO3a attenuates survivin expression (33). It is important to note that ATRA-induced FoxO signalling may promote apoptosis, not only in cells of human sebaceous glands and meibomian glands, but also in neural crest cells, neuroblastoma cells, hippocampal neurones, lymphoma cells, melanoma cells, promyelocytic leukaemia cells, and others (30). Thus, the question arises: Are all adverse effects of isotretinoin caused by apoptosis?

APOPTOSIS AND TERMINAL DIFFERENTIATION

Retinoids modify keratinocyte differentiation and, for this reason, are used in the treatment of a variety of disorders of keratinization, including acne. In primary epidermal keratinocytes, ATRA has been demonstrated to regulate many genes associated with cell cycle control and programmed cell death (34). It should be kept in mind that terminal differentiation of epidermal keratinocytes is finally completed by cornification, which is regarded as a specialized process of keratinocyte apoptosis (35, 36). Apoptosis is also involved in the process of holocrine secretion of sebocytes, as well as telogen induction in hair cycling. Intriguingly, the death ligand TRAIL has recently been demonstrated to be involved in keratinocyte differentiation, which requires caspase activation and p63 expression (37). Thus, apoptosis is a critical process for tissue homeostasis and epidermal differentiation. Apoptosis is the result of increased death signalling but decreased pro-survival signalling. 

The presented hypothesis, supported by translational evidence, confirms that the undesired effects of isotretinoin on non-target cells are mediated by isotretinoin-induced apoptosis.

APOPTOSIS AND TERATOGENICITY

Treatment with isotretinoin during pregnancy is associated with an increased risk of teratogenicity (38). Preventing foetal exposure to isotretinoin is widely acknowledged as an important safety issue. The iPLEDGE programme is the latest in a series of Food and Drug Administration-mandated risk management programmes designed to prevent pregnancies in female patients of childbearing potential who are taking isotretinoin (39). Isotretinoin exerts teratogenic effects in laboratory animals and humans (40). In humans, a characteristic pattern of malformations involves craniofacial, cardiac, thymic and central nervous system structures (40–43). Some of the most characteristic abnormalities include microtia, anotia, micrognathia, conotruncal heart defects and aortic arch abnormalities, thymic ectopia or aplasia, cerebellar vermis agenesis, and various neuronal migration anomalies (40). The major features of the syndrome are explained by the effect of the drug on neural crest cells. During neural development, massive cell death occurs in vertebrates (44). Programmed cell death (apoptosis) is an evolutionarily conserved contributor to nervous system development and requires a well-regulated balance of apoptotic signalling during most critical periods that ensures appropriate cell differentiation and maturation in the developing nervous system (45). Remarkably, increased neural crest cell apoptosis is also the underlying cause in foetal alcohol syndrome (46). ATRA leads to large-scale reprogramming of cranial neural crest gene expression (47), which results in increased apoptosis (48). Animal studies have confirmed that administration of isotretinoin increases apoptosis of neural crest cells (49–51). Excessive cell death, apparently limited to trigeminal ganglion neuroblasts of placodal origin, follows administration of isotretinoin at the time of ganglion formation and leads to malformations (49). In mouse embryos and primary cell cultures, isotretinoin caused significant overall growth retardation, especially in the primary and secondary palatal processes. In embryos explanted on day 10 of gestation and exposed for 24 or 48 h, the mesenchyme beneath the epithelium of the nasal and maxillary processes contained pyknotic nuclei (signs of apoptosis) as well as dramatically reduced numbers of nuclei incorporating 3H-thymidine (50). Increased neural crest cell apoptosis is the crucial mechanism of isotretinoin-induced craniofacial malformations (52). 

Heart defects and aortic arch abnormalities are part of teratogenetic effects of isotretinoin (40–43). Isotretinoin causes a characteristic pattern of heart defects, which result from impaired migration of neural crest cells. Morphogenesis and developmental remodelling of cardiovascular tissues involve coordinated regulation of cell proliferation and apoptosis. In the heart there is clear evidence for focal apoptosis as a contributor to development of the embryonic outflow tract, cardiac valves, conducting system, and the developing coronary vasculature (53). Specific cardiovascular targets of ATRA action include effects on the specification of cardiovascular tissues during early development, such as anteroposterior patterning of the early heart, left/right decisions and cardiac situs and endocardial cushion formation (54–57). Proliferation in heart tissue of whole chick embryo cultures was inhibited in medium with 10–6 M isotretinoin to 62% of the control level in myocardium (58). Taken together, all these observations underline that neural crest cell apoptosis plays the key role in the teratogenicity of isotretinoin.

APOPTOSIS AND DEPRESSION

Recent studies have demonstrated that the hippocampus is one of the brain regions where new neurones are constantly formed; a phenomenon called neurogenesis (59–61). Recent theories for the pathogenesis of depression suggest decreased hippocampal and prefrontal cortex neurogenesis (59–61). In particular, the addition of new neurones within the hippocampus, a limbic region implicated in mood disorders, is compromised in animal models of depression (61). In contrast, antidepressant treatment seems to operate by an increase in neurogenesis, which is chronologically seen during the same period as the clinical improvement (61, 62). Another irregularity in the hippocampus associated with depression is reduction in hippocampal volume. Intriguingly, isotretinoin treatment of mice results in both decreased hippocampal neurogenesis and reduction in hippocampal volume (63, 64). Treatment of hypothalamic cells with 10 ?M isotretinoin for 48 h decreased cell growth to 45.6?±?13% of control (65). Griffin et al. (65) hypothesized that the ability of isotretinoin to decrease hypothalamic cell numbers may contribute to the increased depression-related behaviours observed in mice. A recent study confirmed that intracerebroventricularly applied ATRA to adult rats increased RAR? protein expression in the hippocampus, suggesting an activation of ATRA-induced signalling mechanisms (66). In these rats, ATRA-induced impairments in hippocampal neurogenesis correlated with depression-like symptoms (66). Remarkably, retinoic acid-inducible gene 1 (RAI-1), which increases during neuronal differentiation, was found to be significantly upregulated in brains from patients with schizophrenia, bipolar disorder, or major depression (67). Therefore, isotretinoin-mediated suppression of hippocampal neurogenesis could provide a plausible biological mechanism explaining depressogenic effects of the drug. Individuals with pre-existing impairments of hypothalamic neurogenesis may be at higher risk for the development of isotretinoin-augmented depression. In fact, the literature reviewed from 1960 to June 2010 by Bremner et al. (68) is consistent with isotretinoin-associated depression in a subgroup of vulnerable individuals. The increased risk of isotretinoin-mediated depression is of recent interest in psychiatric research (69–71). Suarez et al. (70) compared isotretinoin-treated vs. non-isotretinoin-treated patients with acne and reported that 13.8% vs. 8.3% of patients developed clinically significant anxiety and/or depression during treatment, respectively. They also concluded that some individuals susceptible to isotretinoin-induced depression do exist, which is in accordance with the author’s own therapeutic experience with oral isotretinoin over more than 30 years. Sundström et al. (72) concluded that an increased risk of attempted suicide was apparent up to 6 months after the end of treatment with isotretinoin. Marron et al. (73) reported a significant reduction in anxiety and depression in isotretinoin-treated patients with acne. However, it is important to realize that acne-mediated depression and isotretinoin-induced depression in a subgroup of patients are 2 different pathogenic entities, which may escape correct detection by epidemiological studies. A subgroup of patients with acne may be more susceptible for isotretinoin-induced neurogenic apoptosis than the great majority of patients who experience an improvement in quality of life after successful acne treatment.

APOPTOSIS AND INFLAMMATORY BOWEL DISEASE

Although the great majority of patients treated with isotretinoin do not experience the development or aggravation of inflammatory bowel disease (IBD) (74–79), an increased risk of IBD in a subgroup of patients cannot be excluded (80–84). It is noteworthy, that a recent study reported excessive death of intestinal epithelial cells (IEC) in the ileal and colonic epithelium to represent a major pathogenetic feature of IBD (85). However, the precise mode of IEC death in IBD has not yet been defined. It is thus conceivable that, in a subgroup of predisposed patients prone to develop IBD, treatment with isotretinoin may aggravate pre-existing IEC apoptosis, thereby promoting the development of IBD, although no scientific evidence is yet available.

APOPTOSIS AND MUCOCUTANEOUS SIDE-EFFECTS

Mucocutaneous adverse effects of oral isotretinoin treatment are dose-dependent and predominantly reflect a decreased production of sebum, reduced thickness of stratum corneum, and altered skin barrier function with increased transepidermal water loss (86). Skin xerosis, especially on exposed skin, and cheilitis are the earliest and the most frequent side-effects that affect almost all treated patients. It should be kept in mind that epidermal keratinocytes undergo a unique form of terminal differentiation and programmed cell death, known as cornification (35, 36). Sebocytes also die in the process of holocrine secretion, which is a less understood process of programmed cell death (35). Notably, ATRA regulates many genes associated with cell cycle control and programmed cell death (34). Genes induced in epidermal keratinocytes by ATRA are the caspases CASP3, CASP6, CASP7 and CASP9 (34). Importantly, ATRA has been shown to induce TRAIL in leukaemia cells (87, 88), which is obviously the therapeutic mode of action of isotretinoin in acute promyelocytic leukaemia and adult T cell leukaemia (89, 90). Wu et al. (37) recently demonstrated that, in keratinocytes, TRAIL is an inducer of both differentiation and apoptosis. Treatment with isotretinoin via activation of TRAIL-induced apoptotic signalling apparently disturbs the homeostatic balance between epidermal cell growth and cell death, which compromises epidermal barrier function (91).

HEPATOCYTE APOPTOSIS

Oral treatment with isotretinoin is often associated with moderate increases in serum levels in alanine aminotransferase (ALT), aspartate aminotransferase (AST), and very low-density lipoproteins (VLDL) triglycerides (92–95). There is good reason to assume that isotretinoin-induced hepatocyte apoptosis explains raised serum transaminase concentrations. Indeed, it has been demonstrated in 2 hepatoma cell lines (Hep3B, HepG2) that ATRA and isotretinoin induce hepatocyte apoptosis (96). However, at present there is no direct evidence that isotretinoin induces hepatocyte apoptosis in treated patients. Upregulated expression of isotretinoin-induced FoxO proteins may explain increased FoxO-mediated expression of hepatic TRAIL (13, 14). In fact, TRAIL has been demonstrated to play a critical role in hepatic cell death and hepatic inflammation (97). Isotretinoin-mediated FoxO1 signalling may also explain isotretinoin-induced hypertriglyceridaemia (30, 98, 99). FoxO1 upregulates microsomal triglyceride transfer protein, which increases hepatic synthesis of triglyceride-rich VLDL (100, 101). Future studies are needed to examine the precise role of upregulated FoxOs in isotretinoin-induced hypertriglyceridaemia.

APOPTOSIS AND MYOPATHY

Muscle-related complaints and increased blood levels of creatine kinase (CK), a specific marker of muscle destruction, have been reported in 16–51% of patients with acne treated with isotretinoin (102–104). Oral isotretinoin-associated adverse muscular effects manifest as myalgia and stiffness and, in rare cases, as true myopathy or rhabdomyolysis. CK has been found to be elevated, occasionally by up to 100 times the normal value, particularly in those patients undergoing vigorous physical exercise (105). Increased TRAIL expression was detected recently in myositis muscle fibres (106). Although direct evidence is missing, it is possible that isotretinoin-induced myalgia may be related to TRAIL-mediated muscle cell apoptosis, explaining CK release during treatment with isotretinoin.

APOPTOSIS AND FERTILITY

Previously, Abali et al. (107) conducted a study about the effect of isotretinoin on ovarian reserve in female Sprague-Dawley rats, which showed a deteriorative ovarian reserve. Notably, the number of ovarian follicles with apoptotic granulosa cells was increased in the experimental groups receiving isotretinoin (107). However, Cinar et al. (108) excluded long-term adverse effects of systemic treatment with isotretinoin on female fertility in humans. 

A recent study found no adverse effects of oral isotretinoin in patients with acne on male fertility, given a total dose of 120 mg/kg over a period of 6 months (109). At present, there is no concern about adverse effects of isotretinoin in male fertility (110). Nevertheless, animal studies demonstrated impaired spermatogenesis. In rats, administration of 2?mg/ml/day isotretinoin for 21 days decreased the number of cyclin D1 and E2F-positive cells (111). In adult male gerbils (Gerbillus cheesemani) isotretinoin induced almost complete cessation of spermatogenesis and produced alterations in the cytoplasm of Leydig cells (112). In the adult lizard Podarcis sicula, administration of ATRA impaired spermatogenesis and enhanced testicular germ cell apoptosis (113). These studies point to an increased risk of isotretinoin-induced germ cell apoptosis in these retinoid-susceptible species.

APOPTOSIS AND HAIR LOSS

Long-term use of isotretinoin in higher doses affects hair growth and is associated with increased hair loss and telogen effluvium in susceptible individuals (86, 114, 115). Hair follicles undergo repetitive stages of cell proliferation and programmed cell death. The catagen stage of physiological apoptosis is connected with dynamic changes in morphology and alterations in gene expression (116, 117). ATRA induces premature hair follicle regression, leading to a catagen-like stage in human hair follicles (118). Hair shaft elongation declined significantly already after 2 days in the ATRA-treated group, and approximately 80% of the ATRA-treated hair follicles had prematurely entered catagen-like stage at day 6, compared with 30% in the control group. This corresponded to an upregulation of apoptotic cells in ATRA-treated hair follicles (116).

APOPTOSIS OF CANCER CELLS

Isotretinoin is the preferred retinoid in the chemoprevention of xeroderma pigmentosum and nevoid basal cell carcinoma syndrome (119). Systemic retinoids play an important role in the treatment of cutaneous T-cell lym­phoma (120). Isotretinoin mediates apoptosis in Dalton’s lymphoma ascites cells (121). Isotretinoin induced growth inhibition and apoptosis in adult T-cell leukaemia (ATL) cells (90, 122). Furthermore, isotretinoin induced apoptosis in B16F-10 melanoma cells (123). Isotretinoin is used in the treatment of acute promyelocytic leukaemia (124, 125). In promyelocytic leukaemia cells, ATRA-induced expression of TRAIL most likely causes leukaemia cell apoptosis (87). The introduction of isotretinoin in the therapy of neuroblastoma, an aggressive childhood tumour, has significantly improved the prognosis of this malignancy (87, 125–127). Notably, neuroblastoma is derived from the peripheral neural crest (125). There is recent evidence that isotretinoin potentiates the apoptotic effects of other drugs, such as melatonin in neuroblastoma cells (128). Thus, isotretinoin-induced apoptosis apparently targets neural crest cells during embryogenesis as well as neural crest cell-derived neuroblastoma cells. Taken together, these data underline that isotretinoin-induced apoptosis is the fundamental chemopreventive and anti-cancer effect of the drug.

CONCLUSION

There is compelling evidence that the major mode of action of isotretinoin is sebocyte apoptosis. Other cells, which are also highly susceptible for isotretinoin-induced apoptosis, such as neural crest cells or neural crest-derived neuroblastoma cells and leukaemia cells, respond with pronounced apoptosis. We have to keep in mind that isotretinoin (13-cis retinoic acid) is the prodrug for cells that are able to isomerize 13-cis- to all-trans retinoic acid (ATRA). The sebocyte is obviously such a cell, which possesses high isomerase activity for the conversion of isotretinoin to ATRA (22). We know that ATRA binds to retinoic acid receptors and thereby modifies the expression of a multitude of genes, including the upregulation of FoxO3a and TRAIL (23–25). At the promoter level, ATRA-induced FoxO3a activates the expression of TRAIL (13, 14) and FoxO1 (129). Finally, TRAIL activates the caspase cascade and orchestrates the programme of apoptosis, whereas FoxO1 induces cell cycle arrest via upregulation of p21 and p27 (30) (Fig. 1). 

Genetic polymorphisms or mutations of components of this death-signalling cascade may explain the observed individual susceptibilities enhancing isotretinoin-mediated apoptosis. Remarkably, genetic polymorphisms in the gene RARA, which encodes RAR?, increase adverse effects of isotretinoin (130). Three-locus haplotype (rs2715554 C/T - rs4890109 G/T - rs9303285 T/C) analysis showed that frequencies of CTG and TTG haplotypes are significantly associated with occurrence of arthralgia, myalgia, nosebleeds and headache in patients treated with isotretinoin. In addition, TCG haplotype was associated with nosebleeds and headache, whereas TTT haplotype was associated with arthralgia and myalgia. Furthermore, levels of AST were increased in patients with the TC genotype of rs2715554 polymorphism, whereas the allele T of rs9303285 was found to be protective against developing depression in patients treated with isotretinoin (130). Thus, genetic variations of critical regulators of isotretinoin-induced apoptotic signalling may explain the increased risk of adverse effects or treatment resistance in subgroups of patients receiving systemic isotretinoin. Notably, activation of caspases, the effectors of the apoptotic machinery, has been detected recently in lymphocytes of patients with depression and anxiety (131). Thus, there is good reason to assume that screening for gene polymorphisms that increase susceptibility for isotretinoin-induced apop­tosis and isotretinoin-associated depression may, in the future, be helpful to identify individuals with increased isotretinoin-mediated apoptotic signalling. At present, depression screening using health questionnaires may be an appropriate method to detect susceptible individuals, who should be closely monitored during treatment with isotretinoin (132). 

In summary, the underlying mechanism of the mode of action and adverse effects of isotretinoin is proposed to be apoptosis (Table I). The magnitude of isotretinoin-induced apoptotic signalling obviously depends on genetic variations, such as RARA polymorphisms. These new insights allow a more balanced and distinguished view on the mode of action of isotretinoin, its risk profile, and explains the enhanced susceptibility for individual apoptosis-related adverse effects in subgroups with genetic variations of isotretinoin-induced apoptotic signalling pathways. 

The author declares no conflicts of interest.


Table I. Isotretinoin-induced apoptosis and drug-mediated effects

REFERENCES
  1. Peck GL, Olsen TG, Yoder FW, Strauss JS, Downing DT, Pandya M, et al. Prolonged remissions of cystic and conglobate acne with 13-cis-retinoic acid. N Engl J Med 1979; 300: 329–333.
    View article    Google Scholar
  2. Honein MA, Lindstrom JA, Kweder SL. Can we ensure the safe use of known human teratogens?: the iPLEDGE test case. Drug Saf 2007; 30: 5–15.
    View article    Google Scholar
  3. Prevost N, English JC. Isotretinoin: update on controversial issues. J Pediatr Adolesc Gynecol 2013; 26: 290–293.
    View article    Google Scholar
  4. Kligman AM, Wheatley VR, Mills OH. Comedogenicity of human sebum. Arch Dermatol 1970; 102: 267–275.
    View article    Google Scholar
  5. Zouboulis CC, Jourdan E, Picardo M. Acne is an inflammatory disease and alterations of sebum composition initiate acne lesions. J Eur Acad Dermatol Venereol 2014; 28: 527–532.
    View article    Google Scholar
  6. Melnik BC. Linking diet to acne metabolomics, inflammation, and comedogenesis: an update. Clin Cosmet Investig Dermatol 2015; 8: 371–388.
    View article    Google Scholar
  7. Strauss JS, Stranieri AM, Farrell LN, Downing DT. The effect of marked inhibition of sebum production with 13 cis-retinoic acid on skin surface lipid composition. J Invest Dermatol 1980; 74: 66–67.
    View article    Google Scholar
  8. Nelson AM, Gilliland KL, Cong Z, Thiboutot DM. 13-cis Retinoic acid induces apoptosis and cell cycle arrest in human SEB-1 sebocytes. J Invest Dermatol 2006; 126: 2178–2189.
    View article    Google Scholar
  9. Nelson AM, Cong Z, Gilliland KL, Thiboutot DM. TRAIL contributes to the apoptotic effect of 13-cis retinoic acid in human sebaceous gland cells. Br J Dermatol 2011; 165: 526–533.
    View article    Google Scholar
  10. Nelson AM, Zhao W, Gilliland KL, Zaenglein AL, Liu W, Thiboutot DM. Neutrophil gelatinase-associated lipocalin mediates 13-cis retinoic acid-induced apoptosis of human sebaceous gland cells. J Clin Invest 2008; 118: 1468–1478.
    View article    Google Scholar
  11. Kelhälä HL, Fyhrquist N, Palatsi R, Lehtimäki S, Väyrynen
    View article    Google Scholar
  12. JP, Kubin ME, et al. Isotretinoin treatment reduces acne lesions but not directly lesional acne inflammation. Exp Dermatol 2016; 25: 477–478.
    View article    Google Scholar
  13. MacFarlane M. TRAIL-induced signalling and apoptosis. Toxicol Lett 2003; 139: 89–97.
    View article    Google Scholar
  14. Zhang X, Tang N, Hadden TJ, Rishi AK. Akt, FoxO and regulation of apoptosis. Biochim Biophys Acta 2011; 1813: 1978–1986.
    View article    Google Scholar
  15. Modur V, Nagarajan R, Evers BM, Milbrandt J. FOXO proteins regulate tumor necrosis factor-related apoptosis inducing ligand expression. Implications for PTEN mutation in prostate cancer. J Biol Chem 2002; 277: 47928–47937.
    View article    Google Scholar
  16. Melnik BC, Zouboulis CC. Potential role of FoxO1 and mTORC1 in the pathogenesis of Western diet-induced acne. Exp Dermatol 2013; 22: 311–315.
    View article    Google Scholar
  17. Mirdamadi Y, Thielitz A, Wiede A, Goihl A, Papakonstantinou E, Hartig R, et al. Insulin and insulin-like growth factor-1 can modulate the phosphoinositide-3-kinase/Akt/FoxO1 pathway in SZ95 sebocytes in vitro. Mol Cell Endocrinol 2015; 415: 32–44.
    View article    Google Scholar
  18. Agamia NF, Abdallah DM, S Sorour O, Morad B, Younan DY. Skin expression of mammalian target of rapamycin (mTOR), forkhead box transcription factor O1 (FoxO1) and serum insulin-like growth factor-1 (IGF-1) in patients with acne vulgaris and their relationship with diet. Br J Dermatol 2016; 174: 1299–1307.
    View article    Google Scholar
  19. Sørensen K, Aksglaede L, Petersen JH, Andersson AM, Juul A. Serum IGF1 and insulin levels in girls with normal and precocious puberty. Eur J Endocrinol 2012; 166: 903–910.
    View article    Google Scholar
  20. Melnik BC. Diet in acne: further evidence for the role of nutrient signalling in acne pathogenesis. Acta Derm Venereol 2012; 92: 228–231.
    View article    Google Scholar
  21. Gross DN, Wan M, Birnbaum MJ. The role of FOXO in the regulation of metabolism. Curr Diab Rep 2009; 9: 208–214.
    View article    Google Scholar
  22. Wang Y, Zhou Y, Graves DT. FOXO transcription factors: their clinical significance and regulation. Biomed Res Int 2014; 2014: 925350.
    View article    Google Scholar
  23. Tsukada M, Schröder M, Roos TC, Chandraratna RA, Reichert U, Merk HF, et al. 13-cis retinoic acid exerts its specific activity on human sebocytes through selective intracellular isomerization to all-trans retinoic acid and binding to retinoid acid receptors. J Invest Dermatol 2000; 115: 321–327.
    View article    Google Scholar
  24. Gudas LJ, Wagner JA. Retinoids regulate stem cell differentiation. J Cell Physiol 2011; 226: 322–330.
    View article    Google Scholar
  25. Kim MJ, Ahn K, Park SH, Kang HJ, Jang BG, Oh SJ, et al. SIRT1 regulates tyrosine hydroxylase expression and differentiation of neuroblastoma cells via FOXO3a. FEBS Lett 2009; 583: 1183–1188.
    View article    Google Scholar
  26. Sakoe Y, Sakoe K, Kirito K, Ozawa K, Komatsu N. FOXO3A as a key molecule for all-trans retinoic acid-induced granulocytic differentiation and apoptosis in acute promyelocytic leukemia. Blood 2010; 115: 3787–3795.
    View article    Google Scholar
  27. Van Der Heide LP, Hoekman MF, Smidt MP. The ins and outs of FoxO shuttling: mechanisms of FoxO translocation and transcriptional regulation. Biochem J 2004; 380: 297–309.
    View article    Google Scholar
  28. Baxter RC. Nuclear actions of insulin-like growth factor binding protein-3. Gene 2015; 569: 7–13.
    View article    Google Scholar
  29. Schedlich LJ, Graham LD, O’Han MK, Muthukaruppan A, Yan X, Firth SM, Baxter RC. Molecular basis of the interaction between IGFBP-3 and retinoid X receptor: role in modulation of RAR-signaling. Arch Biochem Biophys 2007; 465: 359–369.
    View article    Google Scholar
  30. Liu B, Lee HY, Weinzimer SA, Powell DR, Clifford JL, Kurie JM, et al. Direct functional interactions between insulin-like growth factor-binding protein-3 and retinoid X receptor-alpha regulate transcriptional signaling and apoptosis. J Biol Chem 2000; 275: 33607–33613.
    View article    Google Scholar
  31. Melnik BC. Isotretinoin and FoxO1: a scientific hypothesis. Dermatoendocrinol 2011; 3: 141–165.
    View article    Google Scholar
  32. Melnik BC. Pro-inflammatory sebocyte growth and survival signaling in acne vulgaris is reversed by pro-apoptotic isotretinoin signaling. Exp Dermatol 2016; 25: 676–677.
    View article    Google Scholar
  33. Hilmi C, Larribere L, Giuliano S, Bille K, Ortonne JP, Ballotti R, et al. IGF1 promotes resistance to apoptosis in melanoma cells through an increased expression of BCL2, BCL-X(L), and survivin. J Invest Dermatol 2008; 128: 1499–1505.
    View article    Google Scholar
  34. Hilmi C, Larribere L, Deckert M, Rocchi S, Giuliano S, Bille K, et al. Involvement of FKHRL1 in melanoma cell survival and death. Pigment Cell Melanoma Res 2008; 21: 139–146.
    View article    Google Scholar
  35. Lee DD, Stojadinovic O, Krzyzanowska A, Vouthounis C, Blumenberg M, Tomic-Canic M. Retinoid-responsive transcriptional changes in epidermal keratinocytes. J Cell Physiol 2009; 220: 427–439.
    View article    Google Scholar
  36. Eckhart L, Lippens S, Tschachler E, Declercq W. Cell death by cornification. Biochim Biophys Acta 2013; 1833: 3471–3480.
    View article    Google Scholar
  37. Costanzo A, Fausti F, Spallone G, Moretti F, Narcisi A, Botti E. Programmed cell death in the skin. Int J Dev Biol 2015; 59: 73–78.
    View article    Google Scholar
  38. Wu NL, Lee TA, Tsai TL, Lin WW. TRAIL-induced keratinocyte differentiation requires caspase activation and p63 expression. J Invest Dermatol 2011; 131: 874–883.
    View article    Google Scholar
  39. Nau H. Teratogenicity of isotretinoin revisited: species variation and the role of all-trans-retinoic acid. J Am Acad Dermatol 2001; 45: S183–S187.
    View article    Google Scholar
  40. Shin J, Cheetham TC, Wong L, Niu F, Kass E, Yoshinaga MA, et al. The impact of the iPLEDGE program on isotretinoin fetal exposure in an integrated health care system. J Am Acad Dermatol 2011; 65: 1117–1125.
    View article    Google Scholar
  41. Coberly S, Lammer E, Alashari M. Retinoic acid embryopathy: case report and review of literature. Pediatr Pathol Lab Med 1996; 16: 823–836.
    View article    Google Scholar
  42. Lammer EJ, Chen DT, Hoar RM, Agnish ND, Benke PJ, Braun JT, et al. Retinoic acid embryopathy. N Engl J Med 1985; 313: 837–841.
    View article    Google Scholar
  43. Lynburg MC, Khoury MJ, Lammer EJ, Waller KO, Codero JF, Erickson JD. Sensitivity, specificity and positive predictive value of malformations in isotretinoin embryopathy. Teratology 1990; 42: 513–519.
    View article    Google Scholar
  44. Fernhoff PM, Lammer EJ. Craniofacial features of isotretinoin embryopathy. J Pediatr 1984; 5: 595–597.
    View article    Google Scholar
  45. Miura M. Apoptotic and non-apoptotic caspase functions in neural development. Neurochem Res 2011; 36: 1253–1260.
    View article    Google Scholar
  46. Yamaguchi Y, Miura M. Programmed cell death in neurodevelopment. Dev Cell 2015; 32: 478–490.
    View article    Google Scholar
  47. Smith SM, Garic A, Flentke GR, Berres ME. Neural crest development in fetal alcohol syndrome. Birth Defects Res C Embryo Today 2014; 102: 210–220.
    View article    Google Scholar
  48. Williams SS, Mear JP, Liang HC, Potter SS, Aronow BJ, Colbert MC. Large-scale reprogramming of cranial neural crest gene expression by retinoic acid exposure. Physiol Genomics 2004; 19: 184–197.
    View article    Google Scholar
  49. Wang L, Mear JP, Kuan CY, Colbert MC. Retinoic acid induces CDK inhibitors and growth arrest specific (Gas) genes in neural crest cells. Dev Growth Differ 2005; 47: 119–130.
    View article    Google Scholar
  50. Johnston MC, Bronsky PT. Animal models for human craniofacial malformations. J Craniofac Genet Dev Biol 1991; 11: 277–291.
    View article    Google Scholar
  51. Watanabe T, Goulding EH, Pratt RM. Alterations in craniofacial growth induced by isotretinoin (13-cis-retinoic acid) in mouse whole embryo and primary mesenchymal cell culture. J Craniofac Genet Dev Biol 1988; 8: 21–33.
    View article    Google Scholar
  52. Lammer EJ, Armstrong DL. Malformations in hindbrain structures among humans exposed to isotretinoin (13-cis-retinoic acid) during early embryogenesis. In: Morriss-Kay G, editor. Retinoids in Normal Development and Teratogenesis. New York: Oxford University Press, 1991; p. 281–295.
    View article    Google Scholar
  53. Shuler CF. Programmed cell death and cell transformation in craniofacial development. Crit Rev Oral Biol Med 1995; 6: 202–217.
    View article    Google Scholar
  54. Fisher SA, Langille BL, Srivastava D. Apoptosis during cardiovascular development. Circ Res 2000; 87: 856–864.
    View article    Google Scholar
  55. Pan J, Baker KM. Retinoic acid and the heart. Vitam Horm 2007; 75: 257–283.
    View article    Google Scholar
  56. Xavier-Neto J, Rosenthal N, Silva FA, Matos TG, Hochgreb T, Linhares VL. Retinoid signaling and cardiac anteroposterior segmentation. Genesis 2001; 31: 97–104.
    View article    Google Scholar
  57. Niederreither K, Vermot J, Messaddeq N, Schuhbaur B, Chambon P, Dollé P. Embryonic retinoic acid synthesis is essential for heart morphogenesis in the mouse. Development 2001; 128: 1019–1031.
    View article    Google Scholar
  58. Colbert MC. Retinoids and cardiovascular developmental defects. Cardiovasc Toxicol 2002; 2: 25–39.
    View article    Google Scholar
  59. Wiens DJ, Mann TK, Fedderson DE, Rathmell WK, Franck BH. Early heart development in the chick embryo: effects of isotretinoin on cell proliferation, alpha-actin synthesis, and development of contractions. Differentiation 1992; 51: 105–112.
    View article    Google Scholar
  60. Duman RS. Depression: a case of neuronal life and death? Biol Psychiatry 2004; 56: 140–145.
    View article    Google Scholar
  61. Sapolsky RM. Depression, antidepressants and the shrinking hippocampus. Proc Natl Acad Sci USA 2001; 98: 12320–12322.
    View article    Google Scholar
  62. Vaidya VA, Fernandes K, Jha S. Regulation of adult hippocampal neurogenesis: relevance to depression. Expert Rev Neurother 2007; 7: 853–864.
    View article    Google Scholar
  63. Apple DM, Fonseca RS, Kokovay E. The role of adult neurogenesis in psychiatric and cognitive disorders. Brain Res 2016 Jan 19. [Epub ahead of print].
    View article    Google Scholar
  64. Sakai Y, Crandall JE, Brodsky J, McCaffery P. 13-cis retinoic acid (Accutane) suppresses hippocampal cell survival in mice. Ann NY Acad Sci 2004; 1021: 436–440.
    View article    Google Scholar
  65. Crandall JE, Sakai Y, Zhang J, Koul O, Mineur Y, Crusio WE, et al. 13-cis retinoic acid suppresses hippocampal cell division and hippocampal dependent learning in mice. Proc Nat Acad Sci USA 2004; 101: 5111–5116.
    View article    Google Scholar
  66. Griffin JN, Pinali D, Olds K, Lu N, Appleby L, DoanL, et al. 13-cis-Retinoic acid decreases hypothalamic cell number in vitro. Neurosci Res 2010; 68: 185–190.
    View article    Google Scholar
  67. Hu P, Wang Y, Liu J, Meng FT, Qi XR, Chen L, et al. Chronic retinoic acid treatment suppresses adult hippocampal neurogenesis, in close correlation with depressive-like behavior. Hippocampus 2016; 26: 911–923.
    View article    Google Scholar
  68. Haybaeck J, Postruznik M, Miller CL, Dulay JR, Llenos IC, Weis S. Increased expression of retinoic acid-induced gene 1 in the dorsolateral prefrontal cortex in schizophrenia, bipolar disorder, and major depression. Neuropsychiatr Dis Treat 2015; 11: 279–289.
    View article    Google Scholar
  69. Bremner JD, Shearer KD, McCaffery PJ. Retinoic acid and affective disorders: the evidence for an association. J Clin Psychiatry 2012; 73: 37–50.
    View article    Google Scholar
  70. Ludot M, Mouchabac S, Ferreri F. Inter-relationships between isotretinoin treatment and psychiatric disorders: depression, bipolar disorder, anxiety, psychosis and suicide risks. World J Psychiatry 2015; 5: 222–227.
    View article    Google Scholar
  71. Suarez B, Serrano A, Cova Y, Baptista T. Isotretinoin was not associated with depression or anxiety: a twelve-week study. World J Psychiatry 2016; 6: 136–142.
    View article    Google Scholar
  72. Fakour Y, Noormohammadpour P, Ameri H, Ehsani AH, Mokhtari L, Khosrovanmehr N, et al. The effect of isotretinoin (Roaccutane) therapy on depression and quality of life of patients with severe acne. Iran J Psychiatry 2014; 9: 237–240.
    View article    Google Scholar
  73. Sundström A, Alfredsson L, Sjölin-Forsberg G, Gerdén B, Bergman U, Jokinen J. Association of suicide attempts with acne and treatment with isotretinoin: retrospective Swedish cohort study. BMJ 2010; 341: c5812.
    View article    Google Scholar
  74. Marron SE, Tomas-Aragones L, Boira S. Anxiety, depression, quality of life and patient satisfaction in acne patients treated with oral isotretinoin. Acta Derm Venereol 2013; 93: 701–706.
    View article    Google Scholar
  75. Alhusayen RO, Juurlink DN, Mamdani MM, Morrow RL, Shear NH, Dormuth CR; Canadian Drug Safety and Effectiveness Research Network. Isotretinoin use and the risk of inflammatory bowel disease: a population-based cohort study. J Invest Dermatol 2013; 133: 907–912.
    View article    Google Scholar
  76. Rashtak S, Khaleghi S, Pittelkow MR, Larson JJ, Lahr BD, Murray JA. Isotretinoin exposure and risk of inflammatory bowel disease. JAMA Dermatol 2014; 150: 1322–1326.
    View article    Google Scholar
  77. Racine A, Cuerq A, Bijon A, Ricordeau P, Weill A, Allemand H, et al. Isotretinoin and risk of inflammatory bowel disease: a French nationwide study. Am J Gastroenterol 2014; 109: 563–569.
    View article    Google Scholar
  78. Coughlin SS. Clarifying the purported association between isotretinoin and inflammatory bowel disease. J Environ Health Sci 2015; 1 (2). doi: 10.15436/2378-6841.15.007.
    View article    Google Scholar
  79. Lee SY, Jamal MM, Nguyen ET, Bechtold ML, Nguyen DL. Does exposure to isotretinoin increase the risk for the development of inflammatory bowel disease? A meta-analysis. Eur J Gastroenterol Hepatol 2016; 28: 210–216.
    View article    Google Scholar
  80. Lebwohl B, Sundström A, Jabri B, Kupfer SS, Green PH, Ludvigsson JF. Isotretinoin use and celiac disease: a population-based cross-sectional study. Am J Clin Dermatol 2014; 15: 537–542.
    View article    Google Scholar
  81. Passier JL, Srivastava N, van Puijenbroek EP. Isotretinoin-induced inflammatory bowel disease. Neth J Med 2006; 64: 52–54.
    View article    Google Scholar
  82. Reddy D, Siegel CA, Sands BE, Kane S. Possible association between isotretinoin and inflammatory bowel disease. Am J Gastroenterol 2006; 101: 1569–1573.
    View article    Google Scholar
  83. Papageorgiou NP, Altman A, Shoenfeld Y. Inflammatory bowel disease: adverse effect of isotretinoin. Isr Med Assoc J 2009; 11: 505–506.
    View article    Google Scholar
  84. Bharmal R, Anderson SH. Exacerbation of inflammatory bowel disease with isotretinoin. JRSM Short Rep 2010; 1: 58.
    View article    Google Scholar
  85. Papaconstantinou I, Stefanopoulos A, Papailia A, Zeglinas C, Georgopoulos I, Michopoulos S. Isotretinoin and ulcerative colitis: a case report and review of the literature. World J Gastrointest Surg 2014; 6: 142–145.
    View article    Google Scholar
  86. Blander JM. Death in the intestinal epithelium – basic biology and implications for inflammatory bowel disease. FEBS J 2016; 283: 2720–2730.
    View article    Google Scholar
  87. Shalita AR. Mucocutaneous and systemic toxicity of retinoids: monitoring and management. Dermatologica 1987; 175: 151–157.
    View article    Google Scholar
  88. Altucci L, Rossin A, Raffelsberger W, Reitmair A, Chomienne C, Gronemeyer H. Retinoic acid-induced apoptosis in leukemia cells is mediated by paracrine action of tumor-selective death ligand TRAIL. Nat Med 2001; 7: 680–686.
    View article    Google Scholar
  89. Jiménez-Lara AM, Clarke N, Altucci L, Gronemeyer H. Retinoic-acid-induced apoptosis in leukemia cells. Trends Mol Med 2004; 10: 508–515.
    View article    Google Scholar
  90. Bell BA, Findley HW, Krischer J, Whitehead VM, Holbrook T, Haggard ME, et al. Phase II study of 13-cis-retinoic acid in pediatric patients with acute nonlymphocytic leukemia – a Pediatric Oncology Group study. J Immunother 1991; 10: 77–83.
    View article    Google Scholar
  91. Maeda Y, Miyatake J, Sono H, Matsuda M, Tatsumi Y, Horiuchi F, et al. 13-cis Retinoic acid inhibits growth of adult T cell leukemia cells and causes apoptosis; possible new indication for retinoid therapy. Intern Med 1996; 35: 180–184.
    View article    Google Scholar
  92. Lippens S, Denecker G, Ovaere P, Vandenabeele P, Declercq W. Death penalty for keratinocytes: apoptosis versus cornification. Cell Death Differ 2005; 12 Suppl 2: 1497–1508.
    View article    Google Scholar
  93. Vieira AS, Beijamini V, Melchiors AC. The effect of isotretinoin on triglycerides and liver aminotransferases. An Bras Dermatol 2012; 87: 382–387.
    View article    Google Scholar
  94. K?z?lyel O, Metin MS, Elmas ÖF, Çay?r Y, Akta? A. Effects of oral isotretinoin on lipids and liver enzymes in acne patients. Cutis 2014; 94: 234–238.
    View article    Google Scholar
  95. Hansen TJ, Lucking S, Miller JJ, Kirby JS, Thiboutot DM, Zaenglein AL. Standardized laboratory monitoring with use of isotretinoin in acne. J Am Acad Dermatol 2016; 75: 323–328.
    View article    Google Scholar
  96. Lee YH, Scharnitz TP, Muscat J, Chen A, Gupta-Elera G, Kirby JS. Laboratory monitoring during isotretinoin therapy for acne: a systematic review and meta-analysis. JAMA Dermatol 2016; 152: 35–44.
    View article    Google Scholar
  97. Arce F, Gätjens-Boniche O, Vargas E, Valverde B, Díaz C. Apoptotic events induced by naturally occurring retinoids ATRA and 13-cis retinoic acid on human hepatoma cell lines Hep3B and HepG2. Cancer Lett 2005; 229: 271–281.
    View article    Google Scholar
  98. Zheng SJ, Wang P, Tsabary G, Chen YH. Critical roles of TRAIL in hepatic cell death and hepatic inflammation. J Clin Invest 2004; 113: 58–64.
    View article    Google Scholar
  99. Melnik BC, Bros U, Plewig G. Evaluation of the atherogenic risk of isotretinoin-induced and etretinate-induced alterations of lipoprotein cholesterol metabolism. J Invest Dermatol 1987; 88 (3 Suppl): 39s–43s.
    View article    Google Scholar
  100. Bershad S, Rubinstein A, Paterniti JR, Le NA, Poliak SC, Heller B, et al. Changes in plasma lipids and lipoproteins during isotretinoin therapy for acne. N Engl J Med 1985; 313: 981–985.
    View article    Google Scholar
  101. Kamagate A, Qu S, Perdomo G, Su D, Kim DH, Slusher S, Meseck M, et al. FoxO1 mediates insulin-dependent regulation of hepatic VLDL production in mice. J Clin Invest 2008; 118: 2347–2364.
    View article    Google Scholar
  102. Kim DH, Zhang T, Ringquist S, Dong HH. Targeting FoxO1 for hypertriglyceridemia. Curr Drug Targets 2011; 12: 1245–1255.
    View article    Google Scholar
  103. Landau M, Mesterman R, Ophir J, Mevorah B, Alcalay J, Harel A, et al. Clinical significance of markedly elevated serum creatine kinase levels in patients with acne on isotretinoin. Acta Derm Venereol 2001; 81: 350–352.
    View article    Google Scholar
  104. Kaymak Y. Creatine phosphokinase values during isotretinoin treatment for acne. Int J Dermatol 2008; 47: 398–401.
    View article    Google Scholar
  105. Heudes AM, Laroche L. Muscular damage during isotretinoin treatment. Ann Dermatol Venereol 1998; 125: 94–97.
    View article    Google Scholar
  106. Chroni E, Monastirli A, Tsambaos D. Neuromuscular adverse effects associated with systemic retinoid dermato­therapy: monitoring and treatment algorithm for clinicians. Drug Saf 2010; 33: 25–34.
    View article    Google Scholar
  107. Alger HM, Raben N, Pistilli E, Francia DL, Rawat R, Getnet D, et al. The role of TRAIL in mediating autophagy in myositis skeletal muscle: a potential nonimmune mechanism of muscle damage. Arthritis Rheum 2011; 63: 3448–3457.
    View article    Google Scholar
  108. Abali R, Yuksel MA, Aktas C, Celik C, Guzel S, Erfan G, et al. Decreased ovarian reserve in female Sprague-Dawley rats induced by isotretinoin (retinoic acid) exposure. Reprod Biomed Online 2013; 27: 184–191.
    View article    Google Scholar
  109. Cinar SL, Kartal D, Aksoy H, Cinar E, Aydin T, Öz L, et al. Long term effect of systemic isotretinoin on female fertility. Cutan Ocul Toxicol 2016 Jul 5. [Epub ahead of print].
    View article    Google Scholar
  110. Çinar L, Kartal D, Ergin C, Aksoy H, Karadag MA, Aydin T, et al. The effect of systemic isotretinoin on male fertility. Cutan Ocul Toxicol 2016; 35: 296–299.
    View article    Google Scholar
  111. Millsop JW, Heller MM, Eliason MJ, Murase JE. Dermatological medication effects on male fertility. Dermatol Ther 2013; 26: 337–346.
    View article    Google Scholar
  112. Gencoglan G, Tosun M. Effects of isotretinoin on spermatogenesis of rats. Cutan Ocul Toxicol 2011; 30: 55–60.
    View article    Google Scholar
  113. Sadek IA, Abdul-Mohsen MH. Long-term administration of vitamin A and the process of spermatogenesis. East Mediterr Health J 1999; 5: 123–129.
    View article    Google Scholar
  114. Comitato R, Esposito T, Cerbo G, Angelini F, Varriale B, Cardone A. Impairment of spermatogenesis and enhancement of testicular germ cell apoptosis induced by exogenous all-trans-retinoic acid in adult lizard Podarcis sicula. J Exp Zool A Comp Exp Biol 2006; 305: 288–298.
    View article    Google Scholar
  115. Heilgemeir GP, Braun-Falco O, Plewig G, Sund M. Effect of 13-cis-retinoic acid on hair growth. Hautarzt 1982; 33: 533–536.
    View article    Google Scholar
  116. Kmie? ML, Pajor A, Broniarczyk-Dy?a G. Evaluation of biophysical skin parameters and assessment of hair growth in patients with acne treated with isotretinoin. Postepy Dermatol Alergol 2013; 30: 343–349.
    View article    Google Scholar
  117. Botchkareva NV, Ahluwalia G, Shander D. Apoptosis in the hair follicle. J Invest Dermatol 2006; 126: 258–264.
    View article    Google Scholar
  118. Veselá B, Matalová E. Expression of apoptosis-related genes in the mouse skin during the first postnatal catagen stage, focused on localization of Bnip3L and caspase-12. Connect Tissue Res 2015; 56: 326–335.
    View article    Google Scholar
  119. Foitzik K, Spexard T, Nakamura M, Halsner U, Paus R. Towards dissecting the pathogenesis of retinoid-induced hair loss: all-trans retinoic acid induces prematurehair follicle regression (catagen) by upregulation of transforming growth factor-beta2 in the dermal papilla. J Invest Dermatol 2005; 124: 1119–1126.
    View article    Google Scholar
  120. Bettoli V, Zauli S, Virgili A. Retinoids in the chemoprevention of non-melanoma skin cancers: why, when and how. J Dermatolog Treat 2013; 24: 235–237.
    View article    Google Scholar
  121. Huen AO, Kim EJ. The role of systemic retinoids in the treatment of cutaneous T-cell lymphoma. Dermatol Clin 2015; 33: 715–729.
    View article    Google Scholar
  122. Guruvayoorappan C, Pradeep CR, Kuttan G. 13-cis-Retinoic acid mediates apoptosis in Dalton’s lymphoma ascites cells by regulating gene expression. J Basic Clin Physiol Pharmacol 2007; 18: 267–276.
    View article    Google Scholar
  123. Fujimura S, Suzumiya J, Anzai K, Ohkubo K, Hata T, Yamada Y, et al. Retinoic acids induce growth inhibition and apoptosis in adult T-cell leukemia (ATL) cell lines. Leuk Res 1998; 22: 61