Next Article in Journal
Classification and Antioxidant Activity Evaluation of Edible Oils by Using Nanomaterial-Based Electrochemical Sensors
Previous Article in Journal
Discovery of Novel Thiazolidinedione-Derivatives with Multi-Modal Antidiabetic Activities In Vitro and In Silico
Previous Article in Special Issue
Sclerostin Depletion Induces Inflammation in the Bone Marrow of Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Opinion

RANKL-RANK-OPG Pathway in Charcot Diabetic Foot: Pathophysiology and Clinical-Therapeutic Implications

1
Orthopedics and Trauma Surgery Unit, Department of Ageing, Neurosciences, Head-Neck and Orthopedics Sciences, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
2
U.O.C. II Clinic of Orthopaedics and Traumatology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
3
Department of Medicine Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(3), 3014; https://doi.org/10.3390/ijms24033014
Submission received: 26 December 2022 / Revised: 17 January 2023 / Accepted: 1 February 2023 / Published: 3 February 2023
(This article belongs to the Special Issue Advances in Osteoimmunology and Bone Biology)

Abstract

:
Charcot Foot (CF), part of a broader condition known as Charcot Neuro-Osteoarthropathy (CNO), is characterized by neuropathic arthropathy with a progressive alteration of the foot. CNO is one of the most devastating complications in patients with diabetes mellitus and peripheral neuropathy but can also be caused by neurological or infectious diseases. The pathogenesis is multifactorial; many studies have demonstrated the central role of inflammation and the Receptor Activator of NF-κB ligand (RANKL)-Receptor Activator of NF-κB (RANK)-Osteoprotegerin (OPG) pathway in the acute phase of the disease, resulting in the serum overexpression of RANKL. This overexpression and activation of this signal lead to increased osteoclast activity and osteolysis, which is a prelude to bone destruction. The aim of this narrative review is to analyze this signaling pathway in bone remodeling, and in CF in particular, to highlight its clinical aspects and possible therapeutic implications of targeting drugs at different levels of the pathway. Drugs that act at different levels in this pathway are anti-RANKL monoclonal antibodies (Denosumab), bisphosphonates (BP), and calcitonin. The literature review showed encouraging data on treatment with Denosumab, although in a few studies and in small sample sizes. In contrast, BPs have been re-evaluated in recent years in relation to the high possibility of side effects, while calcitonin has shown little efficacy on CNO.

1. Introduction

Charcot Foot (CF), part of a broader condition known as Charcot Neuro-Osteoarthropathy (CNO), is a chronic and degenerative disease characterized by a progressive loss of protective sensibility in the foot and ankle which, through repeated trauma, leads to destruction of bone, joints, and surrounding structures [1]. It is one of the most devastating complications of patients with diabetes and peripheral neuropathy [2].
CNO is related to several neurological, infectious diseases and toxic syndromes such as alcohol abuse [3,4]. In 1868, for the first time, Jean-Martin Charcot described the CF as a complication in patients with tabe dorsalis (myelopathy due to syphilis), whereas in 1936 William Reilly Jordan described foot and ankle CNO as a complication of diabetes for the first time [2]. The joints most affected by CNO are the tarsal, metatarsals, and phalanges, although localizations such as the knee, hip, wrist and spine have also occurred [5,6,7,8].
There are two main classification models of CF: the Modified Eichenholtz classification [9], which relies on clinical and radiographic findings, and the Brodsky classification [10], which focuses on the anatomical distribution of the affected bone segments. The modified Eichenholtz classification ranges from Stage 0 (acute-inflammatory phase), in which the patient presents only cutaneous signs but no visible changes on radiography, to Stage 1 (fragmentation phase), with radiographic evidence of bone destruction, dislocation or subluxation, to Stage 2 (coalescence phase), with fusion of large fragments of adjacent bones and new periosteal bone formation, up to Stage 3 (consolidation phase) with remodeling and new bone formation with possible gross residual deformity [9]. According to Brodsky’s classification, CNO of the foot initially occurs in the Lisfranc joint (tarsometatarsal) and in the tarsus minor (type 1); type 2 occurs in the transverse tarsus (Chopart’s joint), subtalar and peritalar. Type 3a involves the ankle and tibio-talar joints, while type 3b also involves the calcaneus with associated Achilles tendon insufficiency [10].
In the acute phase of CF, generally in afebrile patients with normal vital signs and infection blood markers, the foot is characterized by swelling, warmth and erythema. Usually, skin temperature is increased (from 2 °C to 8 °C) compared to the contralateral; pain may not always be present due to diabetic peripheral neuropathy (this occurs approximately in 50% of cases) and can be associated with impaired deep tendon reflexes, especially the Achilles reflex [11,12].
This phase, often undiagnosed and rapidly progressive, leads to the chronic phase of the disease, with severe deformity, prominences of the bone and a structurally deformed foot [13]. A late and pathognomonic sign is a “rocker bottom foot”, characterized by a prominent calcaneus/heel and a convexly rounded sole [14]. Diagnosis is based on the clinic, often requires radiography, computed tomography (CT) and magnetic resonance imaging (MRI) for any differential diagnosis, especially in the acute and fragmentation phase with bone destruction [15,16,17].
Surgical treatment is reserved for the treatment of deformities and ulcers in the chronic phase; the type of deformity and the patient’s condition will lead to different types of surgical treatment, such as exostectomy, arthrodesis (by internal or external fixation) or amputation [18].
In recent decades, CF is described as a disease with an increased inflammatory response and osteolysis [19], hence many authors have focused on inflammatory and bone remodeling pathways in CNO to better understand their pathogenetic mechanisms and possible therapeutic implications [13,20,21].
The aim of this narrative review is to analyze the Receptor Activator of NF-κB ligand (RANKL)-Receptor Activator of NF-κB (RANK)-Osteoprotegerin (OPG) pathway in CF, highlighting its role in bone remodeling and its correlation with the inflammatory response, and to evaluate the possible therapeutic implications of targeting drugs at different levels of the pathway based on the available literature.

2. RANKL-RANK-OPG Signaling Pathway in Bone Remodeling

Bone remodeling is orchestrated by the activity of osteoblasts (OBs), which produce new bone, and osteoclasts (OCs), that instead reabsorb it. OBs are mononuclear cells derived from mesenchymal stem cell precursors (MSC) [22], and OCs are large multinucleated cells derived from a hematopoietic precursor via the fusion of progenitor cells of mononuclear osteoclasts (OCs) [23].
In 1981, Rodan and Martin hypothesized that OCs genesis was regulated by OBs [24]; while in the mid-1990s it was discovered that this regulation occurs through the expression of members of the Tumor Necrosis Factor (TNF) superfamily, such as the Receptor Activator of NF-κB ligand (RANKL) and Osteoprotegerin (OPG), with the activation of Receptor Activator of NF-κB (RANK) on the cell membrane of the OCs precursors [25].
RANKL-RANK signaling is essential for OCs genesis and differentiation. RANKL, a transmembrane protein resident in the cell membrane of OBs and their precursors, can be released from the membrane by proteolysis from extracellular proteases (disintegrin and/or metalloprotease-7) [26], by activating the RANK receptor located on the cell membrane of the OCs precursors [27] (Figure 1). OPG, through RANKL binding, removes it from interaction with RANK, blocking downstream intracellular signal transduction and thus OCs genesis [28].
Intracellular signal transduction is mediated by the TNF receptor-associated factor 6 (TRAF6) that links the cell surface receptors to downstream kinase cascades, with the activation of transcription factors, such as Nuclear Factor kB (NF-kB) and Activator Protein-1 (AP-1) [29], Mitogen-Activated Protein Kinase (MAPK) and an anti-apoptotic program via c-Src-Akt/Protein Kinase B (PKB) [26]. Therefore, TRAF6 is a crucial factor for OCs formation and activation [30], as shown in a study on mice with inactivating mutation of TRAF6 (TRAF6-/-) with severe osteopetrosis [31].
The recruitment of the family of MAPK leads to the nuclear translocation of c-Fos and c-Jun transcription factors [32], while NF-kB acts as a costimulatory signal for the activation of c-Fos. The importance of c-Fos activation is proven by in vivo studies showing how homozygous c-Fos-/- mutant mice exhibit growth-retarded, severe osteopetrosis and tooth eruption [33].
The transcription factor of c-Fos, together with Nuclear Factor F of Cytoplasmic Activated T cells (NFATc1), triggers the transcription of the genetic program of the OCs genesis. For its activation, NFATc1 requires the intracellular release of calcium ions by phospholipase Cγ2 (PLCγ2). In in-vitro differentiation assays of OCs using embryonic stem cells, cells deficient in NFATc1, showed a defect in the genesis of OCs [34].
Recently, a new RANKL receptor, the leucine-rich G-protein-coupled receptor 4 (LGR4), was discovered that negatively regulates OC differentiation [35]; this receptor enhances bone formation by increasing OB maturation and mineralization, and activates the Wnt/β-catenin pathway [36].
Several studies have demonstrated increased inflammatory activity and bone resorption in CF patients, with evidence of elevated serum levels of inflammatory markers, such as TNF-α, Interleukin-1β (IL-1β) and IL-6, and bone resorption markers, such as RANKL [37,38,39]. Additionally IL-1β and IL-6 play important roles in the pathogenesis by inducing an overproduction of RANKL in CF [37]. In addition, an immunohistochemical analysis of bone samples showed increased OC activity, demonstrated by the presence of a high amount of IL-1, IL-6 and TNF-α [38].
Mabilleau et al. for the first time demonstrated the importance of the (RANKL) pathway in the pathogenesis and treatment of the CF [40]. In the bone, the clinical presentation of serum overexpression of RANKL is an increased activity of OCs, resulting in bone loss; this is also evident in other degenerative diseases, such as osteoporosis, rheumatoid and psoriatic arthritis, in addition to CF [41,42,43]. RANKL is also closely related to bone metastases from tumors such as prostate and breast cancer or multiple myeloma [44]. Breast and prostate cancer cells have been shown not only to express RANK but also to upregulate RANKL expression by OBs and bone marrow stromal cells [45].
A further area of research focused on the allelic locus in CF patients. Burakowska et al. demonstrated that some allelic loci polymorphisms of RANKL and OPG in diabetic patients lead to an increased likelihood of developing CF (in particular OPG 245T/G and OPG 1217C/T are more highly expressed in patients with CNO) [46]. Confirming this, Kloska et al. showed how different alleles coding for OPG and RANKL with different types of monocyte methylation have increased the serum expression in CF (the allelic variants associated with CNO are OPG 245T>G, 1181G>C and 1217C>T and RANKL 290C>T, 643C>T and 693G>C) [47].
Furthermore, an increased RANKL-OPG ratio in the blood is specific for neuropathy and could increase the risk of developing CF disease [46].

3. Therapeutical Implications on RANKL-RANK-OPG Signaling Pathway

The treatment of the acute phase of CF, aimed at resolving the painful symptomatology and controlling the local inflammatory response, is based on non-surgical strategies to reduce the load and edema of the affected foot.
The gold standard of conservative non-pharmacological treatment, based on no weight-bearing and immobilization of the foot and ankle, is the Total Contact Cast (TCC), a cast with padding evenly distributed over the entire limb, but with reinforcement on the tibial crest that is malleolus and around the metatarsal head [11,48,49].
In recent years, the pharmacological treatment of CF has been related to RANKL-RANK-OPG signaling and the OC genesis process, acting at different levels of the pathway [50]. Drugs with direct action on this pathway that have been studied in the literature are anti-RANKL monoclonal antibodies (Denosumab), Bisphosphonates (BP), and Calcitonin [51,52,53,54,55].
Despite the growing interest in this pathway in the pathogenesis of CF, there are few studies in the literature (search via PubMed and Web of Science) on the treatment of CF with these drugs, and these are mostly small-sample studies with a low level of evidence.

3.1. Denosumab

Denosumab is a human monoclonal antibody that selectively binds with high affinity to RANKL, preventing activation of its RANK receptor on the surface of OCs, resulting in the inhibition of any OC activity and the reduction of bone resorption [53,54,55]. Its pharmacokinetics are dose dependent, and the therapeutic effect is achieved with a single subcutaneous dose of 60 mg, which is the standard dose used in all studies reported in the literature on the treatment of CF [55]. Encouraging results have been reported with the use of Denosumab in patients with CF, and it has recently become the main treatment in the refractory active CNO stages of CF [56,57] (Table 1).
  • Busch-Westbroek et al. conducted an observational study of 22 patients to evaluate the efficacy of Denosumab on CF. All patients were treated with no weight-bearing, weekly TCC changes, daily calcium supplementation (500 mg/colecalciferol 800 IU—international units), and subsequent radiographs every 4 weeks. A treatment group of 11 patients received a single subcutaneous dose of 60 mg of Denosumab. At 12 months, the patients in the treatment group showed a decrease in subchondral lysis, an improvement in subchondral bone resurfacing, and a decrease in soft tissue oedema, assessed on conventional radiographs of the affected foot. The TCC time was shorter on average in the treatment group in relation to a faster decrease in 2 °C temperature between the two feet [58].
  • Shofler et al. enrolled seven patients in the acute phase of CF and followed them for one year (with biweekly visits). Patients received a single 60 mg subcutaneous dose of Denosumab and treatment with no weight-bearing and TCC. Efficacy was assessed as the subjects’ exit from the acute phase, defined by normalization of skin temperature by 2 °C relative to the contralateral foot. Patients responded to treatment at an average of 52 days after injection [59].
  • Carvès et al. studied seven patients in the refractory CN stage that were treated with a single subcutaneous dose of 60 mg of denosumab (in case of concomitant osteoporosis, the injection was repeated after 6 months). The follow-up evaluation included clinical, biological examinations and imaging (radiographs and/or glucose analogue (18)F-fluorodeoxyglucose PET-CT). An imaging follow-up was available for five patients and, in four of them, structural damage remained stable on X-ray. PET-CT at baseline was available for all patients, six of whom had increased bone uptake. At the end of treatment, a significant decrease in contrast medium uptake was observed at the joints of the feet. Therefore, denosumab showed a metabolic/anti-inflammatory effect, as measured by 18FDG PET-CT, without adverse events or hypocalcaemia [57].

3.2. Bisphosphonates (BPs)

BPs owe their name to the presence of two phosphonate groups in their chemical structure and constitute a class of drugs with anti-OC action widely used to prevent bone mineral density loss. In recent years, several authors have focused their studies on BPs in CF treatment.
BPs inhibit the action of OCs acting on the enzyme of the cholesterol biosynthesis pathway (farnesyldiphosphate synthase, responsible of geranyl-geranylation, bundling of lipids to regulatory proteins), inhibit their proliferation and shorten their half-life [60]. Another, more recently hypothesized, mechanism of action is that BPs with imidazole groups may be directly involved in the down-regulation of c-Jun and Akt-PKB and the consequent inhibition of c-Fos and NFATc1 expression [61]. Several studies have tested the efficacy of different BPs, initially focusing on pamidronate, later shifting to alendronate and zolendronate (Table 2).
In recent years, the use of BPs has been stigmatized and limited in relation to the clinical corollary of adverse effects reported in the literature, such as the deterioration of renal function, which is particularly important in patients with diabetic neuropathy, and osteonecrosis of the jaw [62]. BPs have shown moderate efficacy in regard to the reduction of bone turnover and the reduction of skin temperature; furthermore, there are as yet no studies confirming efficacy with respect to the reduction of skin deformities and ulcerations [63,64].
  • In 2001, a randomized, double-blind, controlled trial of 39 diabetic patients by Jude et al. compared treatment with a single dose infusion of pamidronate 90 mg versus placebo; the treatment group showed a reduction in bone turnover (measured as a reduction in bone-specific alkaline phosphatase and dehydroxypyridinoline) and, most importantly, a reduction in symptoms related to diabetic neuropathy [65].
  • In a retrospective study, Pakarinen et al. analyzed the medical records and radiographs of 36 feet (32 patients) with acute phase CF. Eighteen received treatment with pamidronate (30–60 mg i.v. once a week for 6 weeks) and a plaster cast without weight-bearing. No significant differences were found in the two groups at the last follow-up [66].
  • In 2005, Pitocco et al. conducted a study of 20 patients in the acute phase of CNO. All patients received a TCC boot for the first 2 months and a pneumatic walker for the other 4 months, then 11 patients were treated with 70 mg alendronate by mouth once a week (test group), and nine control subjects were followed for 6 months. At six months, the authors reported the significant reduction of bone reabsorption markers with increased foot bone density compared with the control group (more evident in the distal phalanxes than in the midfoot) [67].
  • In 2007, in a prospective study of seven patients, Moreno et al. found a rapid resolution of clinical symptoms, with a marked reduction in all markers of bone remodeling and radiological healing at final follow-up following treatment with three doses of pamidronate at 0, 2 and 4 months [68].
  • With the same protocol (three pamidronate 90 mg administrations two months apart), Naqvi et al. showed satisfactory results in terms of resolution of symptoms and ability to walk with load in three cases of CF (two in the subacute phase and 1 in the acute phase) [69].
  • In 2011, a randomized double-blind controlled trial (RCT) by Pakarinen et al. compared zolendronate (4 mg i.v. in three administrations over 3 months) and foot immobilization vs. placebo, in a population of 35 patients. The use of zolendronate has not shown efficacy in the clinical resolution of CNO; rather, patients in the treatment group required a greater number of immobilization days [70].
  • A three-arm double-blind RCT between methylprednisone, zolendronate and placebo showed how the use of cortisone prolonged the time to remission compared to zolendronate and placebo. Inflammatory markers decreased in the three groups, but bone resorption increased in patients treated with methylprednisone, resulting in overall bone loss. Therefore, no benefit was observed in treatment with zolendronate for CF remission [71].
Table 2. Studies on the treatment of CF with Bisphosphonates.
Table 2. Studies on the treatment of CF with Bisphosphonates.
Author, YearPartecipantsTreatmentResults
Jude et al., 2001 [65]39Pamidronate 90 mg (single dose) vs. PlaceboReduction of symptoms related to diabetic neuropathy in treatment group
Pakarinen et al., 2002 [66]32Pamidronate 30–60 mg (i.v., once a week for 6 weeks)No differences between the two groups
Pitocco et al., 2005 [67]20
-
All patients received a TCC boot for the first 2 months and pneumatic walker for the other 4 months
-
11 patients treated with Alendronate 70 mg orally once a week (test group)
-
9 control subjects followed for 6 months
At 6 months, significant reduction of bone reabsorption markers with increased foot bone density compared with the control group (more evident in the distal phalanxes than in the midfoot)
Moreno et al., 2007 [68]7Pamidronate 90 mg (i.v., 3 doses at 2 months-interval)Rapid resolution of symptoms, marked reduction of bone remodeling and radiological healing at final follow-up
Naqvi et al., 2008 [69]3Pamidronate 90 mg (i.v., doses at 2 months-interval)
-
Improvement in swelling, pain, erythema and warmth
-
Patients able to bear weight on their foot and no longer required the use of a walking aid
Pakarinen et al., 2011 [70]35Zolendronate 4 mg (i.v., 3 doses in 3 months) and foot immobilization vs. placebo
-
No significative difference in the two groups
-
The treatment group required more days of immobilization.
Das et al., 2019 [71]36Methylprednisone vs. zolendronate vs. placebo
-
No benefit was observed in treatment with zoledronate for remission of CN
-
Methylprednisone group had a worse prognosis
(mg: milligrams; i.v.: intravenous; TCC: Total Contact Cast).

3.3. Calcitonin

Calcitonin is a polypeptide secreted by the parafollicular C cells of the thyroid gland and acts directly on OCs by inhibiting their bone resorption activity [72]. It is used less in clinical practice than BP and denosumab due to the means of administration (mainly intranasal) [11,49].
  • In a 2006 randomized double-blind study (Table 3), a daily dose of 200 IU calcitonin nasal spray and oral calcium supplementation (treatment group) was compared with oral calcium supplementation alone (control group). Calcitonin treatment showed a good effect on bone turnover at 3 months, with a significant reduction in alkaline phosphatase, but not statistically significant results on diabetic neuropathy control. Foot skin temperature was reduced in both groups, with no significant differences between the two groups [73].

4. Conclusions

Knowledge about the pathogenesis of CF has improved considerably in recent years. Data collected in the literature confirm the central role of the RANKL-RANK-OPG signaling pathway and the importance of this target in the treatment of bone loss and inflammatory signs, therefore research has increased on treatments aimed at interrupting this pathway. Treatment with denosumab has shown encouraging data, although in a few studies and on small sample sizes. In contrast, treatment with BPs has been re-evaluated in recent years in relation to the high possibility of side effects. Calcitonin has shown little efficacy on CNO and is little used for the mode of administration.

Author Contributions

Conceptualization, M.M., N.M. and C.P. (Carlo Perisano); methodology, A.M. and E.T.; software, S.C.; validation, T.G.; formal analysis, S.C.; investigation, A.M.; data curation, C.P. (Chiara Polichetti); writing—original draft preparation, A.M. and C.C.; writing—review and editing, T.G.; visualization, G.M. and C.P. (Carlo Perisano); supervision, G.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare that they have no conflict of interest.

References

  1. Jeffcoate, W.J.; Game, F.; Cavanagh, P.R. The Role of Proinflammatory Cytokines in the Cause of Neuropathic Osteoarthropathy (Acute Charcot Foot) in Diabetes. Lancet 2005, 366, 2058–2061. [Google Scholar] [CrossRef]
  2. Sanders, L.J. The Charcot Foot: Historical Perspective 1827–2003. Diabetes Metab. Res. Rev. 2004, 20 (Suppl. 1), S4–S8. [Google Scholar] [CrossRef] [PubMed]
  3. Mumoli, N.; Camaiti, A. Charcot Foot. CMAJ Can. Med. Assoc. J. 2012, 184, 1392. [Google Scholar] [CrossRef] [PubMed]
  4. Gupta, R. A Short History of Neuropathic Arthropathy. Clin. Orthop. 1993, 296, 43–49. [Google Scholar] [CrossRef]
  5. Phillips, S.; Williams, A.L.; Peters, J.R. Neuropathic Arthropathy of the Spine in Diabetes. Diabetes Care 1995, 18, 867–869. [Google Scholar] [CrossRef] [PubMed]
  6. Berg, E.E. Charcot Arthropathy after Acetabular Fracture. J. Bone Jt. Surg. Br. 1997, 79, 742–745. [Google Scholar] [CrossRef]
  7. Patel, A.; Saini, A.K.; Edmonds, M.E.; Kavarthapu, V. Diabetic Neuropathic Arthropathy of the Knee: Two Case Reports and a Review of the Literature. Case Rep. Orthop. 2018, 2018, 9301496. [Google Scholar] [CrossRef]
  8. Lambert, A.P.; Close, C.F. Charcot Neuroarthropathy of the Wrist in Type 1 Diabetes. Diabetes Care 2005, 28, 984–985. [Google Scholar] [CrossRef]
  9. Shibata, T.; Tada, K.; Hashizume, C. The Results of Arthrodesis of the Ankle for Leprotic Neuroarthropathy. J. Bone Jt. Surg. Am. 1990, 72, 749–756. [Google Scholar] [CrossRef]
  10. Brodsky, J.W.; Rouse, A.M. Exostectomy for Symptomatic Bony Prominences in Diabetic Charcot Feet. Clin. Orthop. Relat. Res. 1993, 296, 21–26. [Google Scholar] [CrossRef]
  11. Marmolejo, V.S.; Arnold, J.F.; Ponticello, M.; Andersen, C.A. Charcot Foot: Clinical Clues, Diagnostic Strategies, and Treatment Principles. Am. Fam. Physician 2018, 97, 594–599. [Google Scholar] [PubMed]
  12. Milne, T.E.; Rogers, J.R.; Kinnear, E.M.; Martin, H.V.; Lazzarini, P.A.; Quinton, T.R.; Boyle, F.M. Developing an Evidence-Based Clinical Pathway for the Assessment, Diagnosis and Management of Acute Charcot Neuro-Arthropathy: A Systematic Review. J. Foot Ankle Res. 2013, 6, 30. [Google Scholar] [CrossRef] [PubMed]
  13. Grant-McDonald, L.; Grant, W. Bone Metabolism in Charcot. Clin. Podiatr. Med. Surg. 2022, 39, 543–557. [Google Scholar] [CrossRef]
  14. Rogers, L.C.; Bevilacqua, N.J. The Diagnosis of Charcot Foot. Clin. Podiatr. Med. Surg. 2008, 25, 43–51. [Google Scholar] [CrossRef] [PubMed]
  15. Mascio, A.; Greco, T.; Maccauro, G.; Perisano, C. Lisfranc Complex Injuries Management and Treatment: Current Knowledge. Int. J. Physiol. Pathophysiol. Pharmacol. 2022, 14, 161–170. [Google Scholar]
  16. Greco, T.; Cianni, L.; De Mauro, D.; Dughiero, G.; Bocchi, M.B.; Cazzato, G.; Ragonesi, G.; Liuzza, F.; Maccauro, G.; Perisano, C. Foot Metastasis: Current Knowledge. Orthop. Rev. 2020, 12, 8671. [Google Scholar] [CrossRef]
  17. Perisano, C.; Greco, T.; Vitiello, R.; Maccauro, G.; Liuzza, F.; Tamburelli, F.C.; Forconi, F. Mueller-Weiss Disease: Review of the Literature. J. Biol. Regul. Homeost. Agents 2018, 32, 157–162. [Google Scholar] [PubMed]
  18. Cianni, L.; Bocchi, M.B.; Vitiello, R.; Greco, T.; De Marco, D.; Masci, G.; Maccauro, G.; Pitocco, D.; Perisano, C. Arthrodesis in the Charcot Foot: A Systematic Review. Orthop. Rev. 2020, 12, 8670. [Google Scholar] [CrossRef]
  19. Jeffcoate, W.J. Theories Concerning the Pathogenesis of the Acute Charcot Foot Suggest Future Therapy. Curr. Diabetes Rep. 2005, 5, 430–435. [Google Scholar] [CrossRef]
  20. Rosskopf, A.B.; Loupatatzis, C.; Pfirrmann, C.W.A.; Böni, T.; Berli, M.C. The Charcot Foot: A Pictorial Review. Insights Imaging 2019, 10, 77. [Google Scholar] [CrossRef]
  21. Pitocco, D.; Scavone, G.; Di Leo, M.; Vitiello, R.; Rizzi, A.; Tartaglione, L.; Costantini, F.; Flex, A.; Galli, M.; Caputo, S.; et al. Charcot Neuroarthropathy: From the Laboratory to the Bedside. Curr. Diabetes Rev. 2019, 16, 62–72. [Google Scholar] [CrossRef] [PubMed]
  22. Castellano, D.; Sepulveda, J.M.; García-Escobar, I.; Rodriguez-Antolín, A.; Sundlöv, A.; Cortes-Funes, H. The Role of RANK-Ligand Inhibition in Cancer: The Story of Denosumab. Oncologist 2011, 16, 136–145. [Google Scholar] [CrossRef]
  23. Sisay, M.; Mengistu, G.; Edessa, D. The RANK/RANKL/OPG System in Tumorigenesis and Metastasis of Cancer Stem Cell: Potential Targets for Anticancer Therapy. OncoTargets Ther. 2017, 10, 3801–3810. [Google Scholar] [CrossRef]
  24. Ducy, P.; Schinke, T.; Karsenty, G. The Osteoblast: A Sophisticated Fibroblast under Central Surveillance. Science 2000, 289, 1501–1504. [Google Scholar] [CrossRef] [PubMed]
  25. Teitelbaum, S.L. Bone Resorption by Osteoclasts. Science 2000, 289, 1504–1508. [Google Scholar] [CrossRef] [PubMed]
  26. Rodan, G.A.; Martin, T.J. Role of Osteoblasts in Hormonal Control of Bone Resorption—A Hypothesis. Calcif. Tissue Int. 1981, 33, 349–351. [Google Scholar] [CrossRef]
  27. Boyce, B.F.; Xing, L. Functions of RANKL/RANK/OPG in Bone Modeling and Remodeling. Arch. Biochem. Biophys. 2008, 473, 139–146. [Google Scholar] [CrossRef]
  28. Nagy, V.; Penninger, J.M. The RANKL-RANK Story. Gerontology 2015, 61, 534–542. [Google Scholar] [CrossRef]
  29. Mp, Y.; Jg, Y. Osteoclastogenesis—Current Knowledge and Future Perspectives. J. Musculoskelet. Neuronal Interact. 2008, 8, 204–216. [Google Scholar]
  30. Udagawa, N.; Koide, M.; Nakamura, M.; Nakamichi, Y.; Yamashita, T.; Uehara, S.; Kobayashi, Y.; Furuya, Y.; Yasuda, H.; Fukuda, C.; et al. Osteoclast Differentiation by RANKL and OPG Signaling Pathways. J. Bone Miner. Metab. 2021, 39, 19–26. [Google Scholar] [CrossRef]
  31. Inoue, J.; Ishida, T.; Tsukamoto, N.; Kobayashi, N.; Naito, A.; Azuma, S.; Yamamoto, T. Tumor Necrosis Factor Receptor-Associated Factor (TRAF) Family: Adapter Proteins that Mediate Cytokine Signaling. Exp. Cell Res. 2000, 254, 14–24. [Google Scholar] [CrossRef] [PubMed]
  32. Wada, T.; Nakashima, T.; Hiroshi, N.; Penninger, J.M. RANKL–RANK Signaling in Osteoclastogenesis and Bone Disease. Trends Mol. Med. 2006, 12, 17–25. [Google Scholar] [CrossRef] [PubMed]
  33. Naito, A.; Azuma, S.; Tanaka, S.; Miyazaki, T.; Takaki, S.; Takatsu, K.; Nakao, K.; Nakamura, K.; Katsuki, M.; Yamamoto, T.; et al. Severe Osteopetrosis, Defective Interleukin-1 Signalling and Lymph Node Organogenesis in TRAF6-Deficient Mice. Genes Cells Devoted Mol. Cell. Mech. 1999, 4, 353–362. [Google Scholar] [CrossRef] [PubMed]
  34. Matsumoto, M.; Sudo, T.; Maruyama, M.; Osada, H.; Tsujimoto, M. Activation of P38 Mitogen-Activated Protein Kinase is Crucial in Osteoclastogenesis Induced by Tumor Necrosis Factor. FEBS Lett. 2000, 486, 23–28. [Google Scholar] [CrossRef] [PubMed]
  35. Grigoriadis, A.E.; Wang, Z.-Q.; Cecchini, M.G.; Hofstetter, W.; Felix, R.; Fleisch, H.A.; Wagner, E.F. C-Fos: A Key Regulator of Osteoclast-Macrophage Lineage Determination and Bone Remodeling. Science 1994, 266, 443–448. [Google Scholar] [CrossRef] [PubMed]
  36. Takayanagi, H.; Kim, S.; Koga, T.; Nishina, H.; Isshiki, M.; Yoshida, H.; Saiura, A.; Isobe, M.; Yokochi, T.; Inoue, J.; et al. Induction and Activation of the Transcription Factor NFATc1 (NFAT2) Integrate RANKL Signaling in Terminal Differentiation of Osteoclasts. Dev. Cell 2002, 3, 889–901. [Google Scholar] [CrossRef]
  37. Connors, J.C.; Hardy, M.A.; Kishman, L.L.; Botek, G.G.; Verdin, C.J.; Rao, N.M.; Kingsley, J.D. Charcot Pathogenesis: A Study of In Vivo Gene Expression. J. Foot Ankle Surg. Off. Publ. Am. Coll. Foot Ankle Surg. 2018, 57, 1067–1072. [Google Scholar] [CrossRef]
  38. Baumhauer, J.F.; O’Keefe, R.J.; Schon, L.C.; Pinzur, M.S. Cytokine-Induced Osteoclastic Bone Resorption in Charcot Arthropathy: An Immunohistochemical Study. Foot Ankle Int. 2006, 27, 797–800. [Google Scholar] [CrossRef]
  39. SaiPrathiba, A.; Senthil, G.; Juttada, U.; Selvaraj, B.; Kumpatla, S.; Viswanathan, V. RANKL Gene Polymorphism as a Potential Biomarker to Identify Acute Charcot Foot Among Indian Population With Type 2 Diabetes: A Preliminary Report. Int. J. Low. Extrem. Wounds 2019, 18, 287–293. [Google Scholar] [CrossRef]
  40. Mabilleau, G.; Petrova, N.L.; Edmonds, M.E.; Sabokbar, A. Increased Osteoclastic Activity in Acute Charcot’s Osteoarthropathy: The Role of Receptor Activator of Nuclear Factor-KappaB Ligand. Diabetologia 2008, 51, 1035–1040. [Google Scholar] [CrossRef]
  41. Tanaka, S.; Tanaka, Y. RANKL as a Therapeutic Target of Rheumatoid Arthritis. J. Bone Miner. Metab. 2021, 39, 106–112. [Google Scholar] [CrossRef] [PubMed]
  42. Jeffcoate, W.J. Charcot Neuro-Osteoarthropathy. Diabetes Metab. Res. Rev. 2008, 24, S62–S65. [Google Scholar] [CrossRef]
  43. Rachner, T.D.; Khosla, S.; Hofbauer, L.C. New Horizons in Osteoporosis. Lancet 2011, 377, 1276–1287. [Google Scholar] [CrossRef]
  44. Luo, J.; Yang, Z.; Ma, Y.; Yue, Z.; Lin, H.; Qu, G.; Huang, J.; Dai, W.; Li, C.; Zheng, C.; et al. LGR4 is a Receptor for RANKL and Negatively Regulates Osteoclast Differentiation and Bone Resorption. Nat. Med. 2016, 22, 539–546. [Google Scholar] [CrossRef]
  45. Carrillo-López, N.; Martínez-Arias, L.; Fernández-Villabrille, S.; Ruiz-Torres, M.P.; Dusso, A.; Cannata-Andía, J.B.; Naves-Díaz, M.; Panizo, S.; On behalf of the European Renal Osteodystrophy (EUROD) Workgroup. Role of the RANK/RANKL/OPG and Wnt/β-Catenin Systems in CKD Bone and Cardiovascular Disorders. Calcif. Tissue Int. 2021, 108, 439–451. [Google Scholar] [CrossRef] [PubMed]
  46. Bruhn-Olszewska, B.; Korzon-Burakowska, A.; Węgrzyn, G.; Jakóbkiewicz-Banecka, J. Prevalence of Polymorphisms in OPG, RANKL and RANK as Potential Markers for Charcot Arthropathy Development. Sci. Rep. 2017, 7, 501. [Google Scholar] [CrossRef]
  47. Kloska, A.; Korzon-Burakowska, A.; Malinowska, M.; Bruhn-Olszewska, B.; Gabig-Cimińska, M.; Jakóbkiewicz-Banecka, J. The Role of Genetic Factors and Monocyte-to-Osteoclast Differentiation in the Pathogenesis of Charcot Neuroarthropathy. Diabetes Res. Clin. Pract. 2020, 166, 108337. [Google Scholar] [CrossRef] [PubMed]
  48. Caputo, G.M.; Ulbrecht, J.; Cavanagh, P.R.; Juliano, P. The Charcot Foot in Diabetes: Six Key Points. Am. Fam. Physician 1998, 57, 2705–2710. [Google Scholar]
  49. Schweitzer, M.; Rockhill, S. Conservative Management of Charcot Neuroarthropathy. Clin. Podiatr. Med. Surg. 2022, 39, 585–594. [Google Scholar] [CrossRef]
  50. Molines, L.; Darmon, P.; Raccah, D. Charcot’s Foot: Newest Findings on Its Pathophysiology, Diagnosis and Treatment. Diabetes Metab. 2010, 36, 251–255. [Google Scholar] [CrossRef]
  51. Guis, S.; Pellissier, J.F.; Arniaud, D.; Turck, F.; Witjas, T.; Roux, H.; Mattei, J.P. Healing of Charcot’s Joint by Pamidronate Infusion. J. Rheumatol. 1999, 26, 1843–1845. [Google Scholar] [PubMed]
  52. Durgia, H.; Sahoo, J.; Kamalanathan, S.; Palui, R.; Sridharan, K.; Raj, H. Role of Bisphosphonates in the Management of Acute Charcot Foot. World J. Diabetes 2018, 9, 115–126. [Google Scholar] [CrossRef]
  53. Baron, R.; Ferrari, S.; Russell, R.G.G. Denosumab and Bisphosphonates: Different Mechanisms of Action and Effects. Bone 2011, 48, 677–692. [Google Scholar] [CrossRef] [PubMed]
  54. Hanley, D.A.; Adachi, J.D.; Bell, A.; Brown, V. Denosumab: Mechanism of Action and Clinical Outcomes. Int. J. Clin. Pract. 2012, 66, 1139–1146. [Google Scholar] [CrossRef]
  55. Deeks, E.D. Denosumab: A Review in Postmenopausal Osteoporosis. Drugs Aging 2018, 35, 163–173. [Google Scholar] [CrossRef]
  56. Chiu, Y.G.; Ritchlin, C.T. Denosumab: Targeting the RANKL Pathway to Treat Rheumatoid Arthritis. Expert Opin. Biol. Ther. 2017, 17, 119–128. [Google Scholar] [CrossRef]
  57. Carvès, S.; Bourgeon-Ghittori, M.; Henry, J.; Belkhir, R.; Besson, F.L.; Levante, S.; Mariette, X.; Seror, R. Denosumab in Active Charcot Neuro-Osteoarthropathy of the Foot. Jt. Bone Spine 2021, 88, 105241. [Google Scholar] [CrossRef] [PubMed]
  58. Busch-Westbroek, T.E.; Delpeut, K.; Balm, R.; Bus, S.A.; Schepers, T.; Peters, E.J.; Smithuis, F.F.; Maas, M.; Nieuwdorp, M. Effect of Single Dose of RANKL Antibody Treatment on Acute Charcot Neuro-Osteoarthropathy of the Foot. Diabetes Care 2017, 41, e21–e22. [Google Scholar] [CrossRef] [PubMed]
  59. Shofler, D.; Hamedani, E.; Seun, J.; Sathananthan, A.; Katsaros, E.; Liggan, L.; Kang, S.; Pham, C. Investigating the Use of Denosumab in the Treatment of Acute Charcot Neuroarthropathy. J. Foot Ankle Surg. Off. Publ. Am. Coll. Foot Ankle Surg. 2021, 60, 354–357. [Google Scholar] [CrossRef] [PubMed]
  60. Reszka, A.A.; Rodan, G.A. Bisphosphonate Mechanism of Action. Curr. Rheumatol. Rep. 2003, 5, 65–74. [Google Scholar] [CrossRef]
  61. Lin, J.; Peng, Y.; Liu, Q.; Li, K.; Lv, G.; Seimbille, Y.; Huang, G.; Gao, F.; Qiu, L. Pharmacological Evaluation of Imidazole-Derived Bisphosphonates on Receptor Activator of Nuclear Factor-κB Ligand-Induced Osteoclast Differentiation and Function. Chem. Biol. Drug Des. 2021, 97, 121–133. [Google Scholar] [CrossRef]
  62. Pazianas, M.; Miller, P.; Blumentals, W.A.; Bernal, M.; Kothawala, P. A Review of the Literature on Osteonecrosis of the Jaw in Patients with Osteoporosis Treated with Oral Bisphosphonates: Prevalence, Risk Factors, and Clinical Characteristics. Clin. Ther. 2007, 29, 1548–1558. [Google Scholar] [CrossRef]
  63. Richard, J.-L.; Almasri, M.; Schuldiner, S. Treatment of Acute Charcot Foot with Bisphosphonates: A Systematic Review of the Literature. Diabetologia 2012, 55, 1258–1264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Game, F.L.; Catlow, R.; Jones, G.R.; Edmonds, M.E.; Jude, E.B.; Rayman, G.; Jeffcoate, W.J. Audit of Acute Charcot’s Disease in the UK: The CDUK Study. Diabetologia 2012, 55, 32–35. [Google Scholar] [CrossRef] [PubMed]
  65. Jude, E.B.; Selby, P.L.; Burgess, J.; Lilleystone, P.; Mawer, E.B.; Page, S.R.; Donohoe, M.; Foster, A.V.; Edmonds, M.E.; Boulton, A.J. Bisphosphonates in the Treatment of Charcot Neuroarthropathy: A Double-Blind Randomised Controlled Trial. Diabetologia 2001, 44, 2032–2037. [Google Scholar] [CrossRef]
  66. Pakarinen, T.-K.; Laine, H.-J.; Honkonen, S.E.; Peltonen, J.; Oksala, H.; Lahtela, J. Charcot Arthropathy of the Diabetic Foot. Current Concepts and Review of 36 Cases. Scand. J. Surg. 2002, 91, 195–201. [Google Scholar] [CrossRef]
  67. Pitocco, D.; Ruotolo, V.; Caputo, S.; Mancini, L.; Collina, C.M.; Manto, A.; Caradonna, P.; Ghirlanda, G. Six-Month Treatment with Alendronate in Acute Charcot Neuroarthropathy: A Randomized Controlled Trial. Diabetes Care 2005, 28, 1214–1215. [Google Scholar] [CrossRef]
  68. Moreno, M.; Gratacós, J.; Casado, E.; Galisteo, C.; Orellana, C.; Larrosa, M. Usefulness of Pamidronate in the Treatment of Charcot’s Arthropathy. Reumatol. Clin. 2007, 3, 257–261. [Google Scholar] [CrossRef]
  69. Naqvi, A.; Cuchacovich, R.; Saketkoo, L.; Espinoza, L.R. Acute Charcot Arthropathy Successfully Treated with Pamidronate: Long-Term Follow-Up. Am. J. Med. Sci. 2008, 335, 145–148. [Google Scholar] [CrossRef] [PubMed]
  70. Pakarinen, T.-K.; Laine, H.-J.; Mäenpää, H.; Mattila, P.; Lahtela, J. The Effect of Zoledronic Acid on the Clinical Resolution of Charcot Neuroarthropathy. Diabetes Care 2011, 34, 1514–1516. [Google Scholar] [CrossRef]
  71. Das, L.; Bhansali, A.; Prakash, M.; Jude, E.B.; Rastogi, A. Effect of Methylprednisolone or Zoledronic Acid on Resolution of Active Charcot Neuroarthropathy in Diabetes: A Randomized, Double-Blind, Placebo-Controlled Study. Diabetes Care 2019, 42, e185–e186. [Google Scholar] [CrossRef] [PubMed]
  72. Wu, H.; Lin, X.-Q.; Long, Y.; Wang, J. Calcitonin Gene-Related Peptide is Potential Therapeutic Target of Osteoporosis. Heliyon 2022, 8, e12288. [Google Scholar] [CrossRef] [PubMed]
  73. Bem, R.; Jirkovská, A.; Fejfarová, V.; Skibová, J.; Jude, E.B. Intranasal Calcitonin in the Treatment of Acute Charcot Neuroosteoarthropathy: A Randomized Controlled Trial. Diabetes Care 2006, 29, 1392–1394. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. RANKL-RANK-OPG signaling pathway.
Figure 1. RANKL-RANK-OPG signaling pathway.
Ijms 24 03014 g001
Table 1. Studies on the treatment of CF with Denosumab.
Table 1. Studies on the treatment of CF with Denosumab.
Author, YearParticipantsTreatmentResults
Busch-Westbroek et al., 2017 [58]22Denosumab 60 mg (single subcutaneous dose) and TCC protocolFracture resolution and shorter TCC treatment time
Shofler et al., 2021 [59]7Denosumab 60 mg (single subcutaneous dose) in the CF acute phaseExit from the acute phase in an average of 52.00 ± 17.89 days after injection
Carvés et al., 2021 [57]7Denosumab 60 mg (single subcutaneous dose) in refractory stage
-
All patients clinically improved
-
5 patients showed stability of structural damage (radiography)
-
4 patients with significant decrease of metabolic activity (PET-CT)
-
No adverse event or hypocalcemia was observed.
(mg: milligrams; TCC: Total Contact Cast).
Table 3. Studies on the treatment of CF with calcitonin.
Table 3. Studies on the treatment of CF with calcitonin.
Author, YearPartecipantsTreatmentResults
Bem et al., 2006 [73]32Calcitonin spray 200 IU + calcium vs. calcium alone
-
Reduced bone turnover at 3 months
-
No significative difference between the two groups in foot-temperature reduction
(IU: International Units).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Greco, T.; Mascio, A.; Comisi, C.; Polichetti, C.; Caravelli, S.; Mosca, M.; Mondanelli, N.; Troiano, E.; Maccauro, G.; Perisano, C. RANKL-RANK-OPG Pathway in Charcot Diabetic Foot: Pathophysiology and Clinical-Therapeutic Implications. Int. J. Mol. Sci. 2023, 24, 3014. https://doi.org/10.3390/ijms24033014

AMA Style

Greco T, Mascio A, Comisi C, Polichetti C, Caravelli S, Mosca M, Mondanelli N, Troiano E, Maccauro G, Perisano C. RANKL-RANK-OPG Pathway in Charcot Diabetic Foot: Pathophysiology and Clinical-Therapeutic Implications. International Journal of Molecular Sciences. 2023; 24(3):3014. https://doi.org/10.3390/ijms24033014

Chicago/Turabian Style

Greco, Tommaso, Antonio Mascio, Chiara Comisi, Chiara Polichetti, Silvio Caravelli, Massimiliano Mosca, Nicola Mondanelli, Elisa Troiano, Giulio Maccauro, and Carlo Perisano. 2023. "RANKL-RANK-OPG Pathway in Charcot Diabetic Foot: Pathophysiology and Clinical-Therapeutic Implications" International Journal of Molecular Sciences 24, no. 3: 3014. https://doi.org/10.3390/ijms24033014

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop