Colchicine is a plant-derived alkaloid that disrupts the cell microtubule system and accumulates in neutrophils, inhibiting neutrophil adhesion and recruitment. Colchicine has been used extensively in the prevention and treatment of gouty arthritis attacks, familial Mediterranean fever attacks and resultant AA amyloidosis, and recurrent pericarditis. Colchicine also disrupts the intracellular traffic of additional inflammatory and fibrosis mediators. Renal fibrosis is the final common pathway of chronic renal disease. Colchicine had anti-fibrotic effects in experimental diabetic nephropathy, renal mass reduction, and cyclosporine nephrotoxicity among others and is undergoing clinical trials for non-diabetic metabolic syndrome and diabetic nephropathy. In this review, we summarize the anti-inflammatory and anti-fibrotic properties of colchicine in experimental and clinical studies in renal diseases or other fibrotic disease processes with renal consequences. We also discuss the potential future uses of colchicine in renal medicine and challenges faced with its use in patients with impaired kidney function.

Colchicine is a plant-derived (colchicum autumnale) alkaloid that has been used for thousands of years to treat nonspecific arthritis [1]. Extensive clinical experience found colchicine safe and efficacious in treating gout and familial Mediterranean fever (FMF). Colchicine has additionally been used in the treatment of Behcet's disease, recurrent pericarditis, and some fibrotic disorders and is undergoing clinical trials for ischemic heart disease and arrhythmia [2]. The most recent area where colchicine has been used is in the field of cardiovascular disease. A recent meta-analysis demonstrated that in different populations of patients with established cardiovascular disease, colchicine reduced the composite cardiovascular outcome by almost 60% and showed a trend toward lower all-cause mortality in patients with coronary artery disease, acute coronary syndrome or stroke, post-angioplasty, or congestive heart failure [3].

Colchicine interferes with microtubule polymerization, thus interfering with the neutrophil function and having an anti-inflammatory effect [4]. In addition, colchicine has anti-fibrotic properties via a number of distinct mechanisms [5]. Since fibrosis is a ubiquitous final pathway in various kidney diseases, it is not surprising that experimental studies have shown favorable effects of colchicine in reducing fibrosis and in delaying the progression of kidney disease. Despite available and promising experimental evidence, scant clinical data regarding colchicine use in renal disease are available.

In this review, we discuss the mechanisms of the anti-fibrotic effect of colchicine in the context of renal disease. We summarize clinical and experimental data regarding colchicine efficacy and safety in renal diseases or in disease processes that may cause kidney injury. We also discuss the potential sources of underutilization as well as some putative indications in which colchicine may be effective.

Alpha and β-tubulin heterodimers constitute microtubules that elongate and contract, thereby altering the structure and function of the cytoskeleton [4]. Microtubules play a role in cell division as well as in many other physiologic processes including signal transduction, cell migration, and secretion [6]. The main mechanism of the action of colchicine is disruption of this microtubule system by binding to tubulins and blocking the assembly and polymerization of microtubules. Low concentrations arrest microtubule growth, whereas higher concentrations promote microtubule depolymerization [4]. Colchicine concentrates avidly in leukocytes, interfering with neutrophil adhesion, recruitment, and activation via microtubule depolymerization [7,8]. Thus, colchicine decreases the release of superoxide anion from activated neutrophils and tyrosine phosphorylation in proteins involved in neutrophil responses to monosodium urate (MSU) and calcium pyrophosphate dehydrate crystals [9]. Not much data is available on colchicine actions on other immune functions. A detailed discussion of the anti-inflammatory effects of colchicine is beyond the scope of this paper and the reader is referred to reviews elsewhere [2,6]. As examples, colchicine suppresses MSU crystal-induced NALP3 (NLRP3) inflammasome activation through the disruption of the intracellular localization of NALP3, thus preventing caspase 1 activation and release of interleukin (IL)-1β and IL-18 [10], downregulates lipopolysaccharide-induced tumor necrosis factor-alpha (TNF-α) secretion by liver macrophages [11] and prevents MSU-induced downregulation of the anti-inflammatory neutrophil receptor myeloid inhibitory C-type lectin-like receptor, thus attenuating inflammation [12].

After oral administration, colchicine is readily absorbed in the jejunum and ileum. Because of its lipophilic structure, it rapidly enters multiple cell types. It is metabolized by the intestinal and hepatic cytochrome P450 (CYP3A4) enzyme systems. Colchicine is a substrate for P-glycoprotein 1 (multidrug resistance protein 1) efflux pump and is predominantly eliminated by hepatobiliary excretion, with gastrointestinal tract lining cell turnover playing a role in its elimination through P-glycoprotein-1. Renal excretion accounts only for 10-20% of total colchicine elimination in patients with normal renal function [13].

Fibrosis is a final common phenomenon in chronic nephropathies, irrespective of their etiology, and is characterized by glomerulosclerosis and/or tubulointerstitial fibrosis [14]. The process is active and much more complex than once thought. The key feature is the accumulation of fibroblasts and extracellular matrix (ECM). Fibroblasts are activated into myofibroblasts via a variety of mechanical factors and fibrogenic cytokines, such as transforming growth factor (TGF)-β1 [15,16]. Therefore, it is plausible to think that preventing or ameliorating fibrosis may halt the progression of chronic kidney disease (CKD).

Colchicine has been used in a number of fibrogenic disease processes, particularly in hepatic fibrosis. The effects of colchicine on histological grade of hepatic fibrosis were explored in a limited number of studies. In a multicenter randomized placebo-controlled trial of patients with primary biliary cirrhosis, colchicine reduced the histological grading score when added to ursodeoxycolic acid [17]. However, a meta-analysis of randomized controlled studies did not reveal an overall remarkable benefit of colchicine in primary biliary cirrhosis [18]. As for the kidney, beneficial effects of colchicine have been observed in a number of primary fibrotic conditions of the kidney as well as in fibrotic processes in which the kidney is involved secondarily (Table 1).

Table 1

Experimental and clinical studies of colchicine in the renal disease processes with prominent fibrosis or in fibrotic disorders in which kidney is involved collaterally

Experimental and clinical studies of colchicine in the renal disease processes with prominent fibrosis or in fibrotic disorders in which kidney is involved collaterally
Experimental and clinical studies of colchicine in the renal disease processes with prominent fibrosis or in fibrotic disorders in which kidney is involved collaterally

Colchicine may curb fibrosis through anti-inflammatory and direct anti-fibrotic actions (Fig. 1).

Fig. 1

Schematic illustration of anti-fibrotic effects of colchicine. Colchicine inhibits the secretion of TGF-β, connective tissue growth factor, and PDGF from leukocytes. These cytokines play a role in the transformation of resting fibroblasts to myofibroblasts, which are capable of secreting copious amounts of ECM. Colchicine also decreases myofibroblast proliferation and ECM synthesis and prevents the release or cell surface expression of inflammatory mediators such as ICAM-1, TNF-α, and IL-1β. CTGF, connective tissue growth factor; ECM, extracellular matrix; EMT, epithelial to mesenchymal transition; ICAM-1, intercellular adhesion molecule; MCP-1, monocyte chemoattractant protein; PDGF, platelet-derived growth factor; TGF-β, transforming growth factor; TNF-α, tumor necrosis factor.

Fig. 1

Schematic illustration of anti-fibrotic effects of colchicine. Colchicine inhibits the secretion of TGF-β, connective tissue growth factor, and PDGF from leukocytes. These cytokines play a role in the transformation of resting fibroblasts to myofibroblasts, which are capable of secreting copious amounts of ECM. Colchicine also decreases myofibroblast proliferation and ECM synthesis and prevents the release or cell surface expression of inflammatory mediators such as ICAM-1, TNF-α, and IL-1β. CTGF, connective tissue growth factor; ECM, extracellular matrix; EMT, epithelial to mesenchymal transition; ICAM-1, intercellular adhesion molecule; MCP-1, monocyte chemoattractant protein; PDGF, platelet-derived growth factor; TGF-β, transforming growth factor; TNF-α, tumor necrosis factor.

Close modal

Lymphocytes and macrophages contribute to renal fibrosis [19,20]. Colchicine inhibits lymphocyte proliferation and function [21,22]. In the rat chronic cyclosporine (CsA) nephrotoxicity model, colchicine prevented macrophage influx and renal tubulointerstitial fibrosis [23]. Similarly, colchicine prevented macrophage infiltration and ECM deposition in rat diabetic nephropathy [24].

High TGF-β expression is a hallmark of and a key contributor to renal fibrosis [25,26,27]. Colchicine suppressed TGF-β1 mRNA expression, TGF-β1 secretion, and collagen synthesis, processing and release in cultured renal fibroblasts [28,29,30]. Colchicine also inhibited the release of fibronectin and fibroblast proliferation, and stimulated tissue collagenase activity [29,31,32].

Matrix metalloproteinases (MMPs) and their inhibitors (TIMPs) degrade ECM proteins. Colchicine alters the expression of various MMPs and TIMPs including MMP-I and TIMPs [29,33].

Epithelial-mesenchymal transition (EMT) has been implicated in renal fibrogenesis [34]. Epithelial cells undergo a morphologic change characterized by the loss of apico-basal polarity and epithelial intercelluler contacts, with increased motility and contractility [14]. In endothelial to mesenchymal transition, endothelial cells acquire fibroblastic properties and may contribute to the accumulation of activated fibroblasts and myofibroblasts in areas of kidney fibrosis [5,35,36]. Colchicine modulates epithelial cell migration [37,38], ECM synthesis, and fibroblast functions [39]. The precise targets of colchicine in EMT remain to be elucidated.

Ozdemir et al. [40] extended these observations to human subjects. In a case-control study of 25 renal transplant recipients with amyloidosis secondary to FMF, allograft biopsies were performed during the first, second, and third years post-transplant. Colchicine use was associated with milder interstitial fibrosis in renal allografts of amyloidotic patients compared with nonamyloidotic controls who did not receive colchicine.

Globally diabetes mellitus is the most common cause of CKD [41] and is characterized by kidney inflammation and ECM accumulation [42]. Functional preclinical studies and early clinical trials suggest that inflammation and macrophage infiltration are key drivers of DN progression [43,44]. Thus, the expression of adhesion molecules such as intercellular adhesion molecule-1 (ICAM-1) is increased in DN [45,46]. Indeed, ICAM-1 deficiency protected mice from DN, reducing macrophage infiltration, glomerular TGF-β1, and fibrosis [47,48]. The renal monocyte chemoattractant protein-1 (MCP-1; CCL2) is also involved in the migration of the macrophage into the diabetic kidney [49]. Colchicine reduced albuminuria, kidney MCP-1 and ICAM-1 expression, inflammatory cell infiltration (mainly monocytes and macrophages), and ECM accumulation in type 1 rat DN [24]. Furthermore, colchicine prevented the diabetes-induced podocyte depletion in rats [24]. This observation is of paramount importance, since podocyte depletion is a central phenomenon in CKD progression. However, human data are needed to confirm whether colchicine improves histopathologic changes. In this regard, an ongoing clinical trial (NCT02035891) is exploring whether low-dose (0.5 mg/day) colchicine reduces microalbuminuria within 18-36 months when compared with placebo in patients with type 2 diabetes and microalbuminuria who have been receiving stable treatment of angiotensin-converting enzyme inhibitor/angiotensin II receptor blocker for at least 3 months. The estimated primary completion date is of June 2018.

Neutrophil dysfunction in diabetic patients contributes to their increased susceptibility to infections and may contribute to systemic inflammation and DN, especially in patients with concomitant CKD. Spontaneous adherence and hydrogen peroxide synthesis of neutrophilic polymorphonuclear leukocytes are significantly higher in neutrophils from diabetic patients with overt proteinuria than from normoalbuminuric diabetic patients or from healthy controls [50,51]. Neutrophils are activated in type-1 diabetes mellitus [52]. Leukocyte activation markers, CD11B on monocytes, and CD11B and CD66B on neutrophils, were higher in diabetic patients than in controls [53]. Neutrophils from type 2 diabetic patients release higher amounts of inflammatory cytokines under spontaneous and LPS-stimulated conditions, than non-diabetics [54]. In fact, neutrophils were more responsive to diabetic conditions than monocytes [54]. Taken together, neutrophil overactivity may contribute to tissue injury and DN, especially when end-organ damage becomes apparent. Thus, colchicine, by decreasing neutrophil pro-inflammatory responses has the potential to ameliorate detrimental effects of activated neutrophils in diabetic subjects.

Renal mass reduction favors the progressive loss of renal function. In a rat remnant kidney model, colchicine inhibited glomerular RhoA activation and attenuated interstitial fibrosis and glomerular sclerosis independently of systemic blood pressure. Colchicine also reduced the pro-fibrotic cytokines TGF-β activity and connective tissue growth factor and the upregulation of ECM proteins, collagen I, and fibronectin. Besides this, colchicine decreased the renal infiltration of lymphocytes and macrophages [55]. RhoA/Rho-kinase signaling is dependent on an intact microtubule network [56] and RhoA kinase inhibition by colchicine could underlie the nephroprotective effect in renal mass reduction [57].

During the 1990s, experimental studies showed potent immunomodulatory actions and beneficial effects of colchicine in renal transplantation. However, despite promising initial results, clinical studies did not follow, possibly due to the advent of newer potent immunosuppressive drugs and reluctance to use colchicine with concerns of systemic toxicity.

Ostermann et al. [21] first showed that colchicine prevented allograft rejection in rats. Colchicine 40 µg/kg intraperitoneally starting 2 h prior to transplantation and continuing at 40 or 10 µg/kg/day resulted in the long-term survival of the allograft compared with controls. Chronic administration of colchicine led to systemic unresponsiveness as evaluated by T cells functional status in a mixed lymphocyte response (MLR) assay. Colchicine inhibition of MLR was dose-dependent [58]. Colchicine also inhibited the generation of cytotoxic T lymphocytes and the cytotoxic T-cell effector function in vitro and decreased the mononuclear cell infiltrates, IgM, C3, and fibrin deposition and the upregulation of activation markers, including IL-2 receptor, MHC class II, and ICAM-I in grafts, suggesting the selective inhibition of Th1 and the sparing of Th2 function. Colchicine also downregulated L-selectin in a dose-dependent manner and inhibited lymphocyte function in vitro [22]. In addition, colchicine prevented the interferon-gamma-induced expression of class II MHC molecules on the surface of colon cancer cells in culture [59]. The early phase of the immune response requires the expression of class II MHC molecules on the surface of antigen-presenting cells. Other contributory mechanisms include the decrease of TNF-α [60] and blockage of IL-2R expression on the surface of peripheral blood mononuclear cells [61]. Thus, colchicine downregulates the alloimmune response in vitro and prevents acute rejection and prolongs graft survival by blocking microtubule-dependent traffic of molecules to the cell surface of T cells and endothelial cells.

Research on colchicine in transplantation did not proceed to human studies. However, chronic allograft nephropathy (CAN) remains a major limitation of modern transplantation medicine [62]. Immunosuppressive drugs, particularly calcineurin inhibitors, contribute to CAN. Thus, the use of additional immunomodulatory drugs such as colchicine may help reduce the dose of calcineurin inhibitors to prevent both rejection episodes and CAN. In addition, colchicine may also be beneficial in CAN through its anti-fibrotic and anti-inflammatory actions [62]. A careful analysis of FMF kidney graft recipients currently on colchicine may provide further insights in this regard.

Calcineurin inhibitors constitute the backbone of modern immunosuppressive regimens for kidney transplantation [63]. However, the protracted use of calcineurin inhibitors may promote chronic graft dysfunction [64]. Colchicine improved experimental CsA nephrotoxicity, decreasing malonyldialdehyde serum levels, TGF-β expression, renal cell apoptosis, tubular atrophy, and interstitial fibrosis [65,66].

Renal amyloidosis is a cause of end-stage renal disease [67,68]. Over 20 structurally different proteins are known to cause amyloidosis [69]. However, 2 major forms are most related to kidney disease, namely, AL and AA amyloidoses.

The fibrils in AL amyloidosis consist of the fragments of the variable portion of monoclonal light chains [70]. Current therapy involves chemotherapy aimed at eliminating the plasma cell clone. High-dose intravenous melphalan followed by autologous stem cell transplantation has emerged as the most effective scheme [71]. In the 1980s and 1990s, colchicine was explored as a single therapy or as an add-on to chemotherapeutic regimens [72,73,74,75,76]. However, addition of colchicine did not provide a significant survival advantage over melphalan and prednisolone alone: in 220 patients with biopsy-proved AL amyloidosis, median survival was 8.5 months for colchicine, 18 months for melphalan and prednisone, and 17 months for melphalan, prednisone, and colchicine.

AA amyloidosis is a disorder characterized by the abnormal systemic deposition of an acute phase reactant, serum amyloid A, in chronic inflammatory diseases. It usually results from FMF and some rheumatologic diseases or chronic infections [77]. Colchicine is used to prevent AA amyloidosis in patients with FMF. When started early and used at sufficient doses with a good compliance, development of AA amyloidosis can effectively be prevented in these patients [78]. Established AA renal amyloidosis is much less responsive to colchicine treatment [79].

The presence of AA amyloidosis does not preclude renal transplantation, but disease recurrence may lead to allograft loss if left untreated. Colchicine has been used effectively to prevent recurrent allograft dysfunction in amyloidotic patients with FMF who underwent renal transplantation. A retrospective study by Livneh et al. [80] revealed that colchicine treatment with a dose of at least 1.5 mg/day effectively prevented recurrent amyloidosis in the allograft kidney. Subsequent retrospective studies confirmed [81,82,83] that in patients maintained on adequate colchicine treatment, renal outcomes of amyloidotic patients were not different from those without amyloidosis. Unverdi et al. [84] reported that the beneficial effect of colchicine therapy was not evident in patients with AA amyloidosis secondary to rheumatologic diseases.

Although tested in a limited number of nephropathies, the anti-inflammatory, anti-fibrotic, and anti-proliferative properties of colchicine suggest that it may protect the kidney in additional disorders, including autosomal dominant polycystic kidney disease (ADPKD) and focal and segmental glomerulosclerosis (FSGS).

Previously we hypothesized that colchicine may be useful in ADPKD [85]. A number of factors have been implicated in the pathophysiology of ADPKD: increased apoptosis, unopposed proliferation of tubule cells, impaired polarization and planar cell polarity, impaired cAMP pathway, ciliary dysfunction, activated mammalian target of rapamycin pathway, and increased TNF-α production [85]. By limiting the proliferation of cyst-lining cells and inflammatory and fibrotic responses, colchicine may improve outcomes in ADPKD and should certainly be tested in experimental animal models.

FSGS is characterized by glomerular fibrosis and loss of podocytes and inflammation contributes to the process [86]. Preservation of podocytes [24] and the anti-inflammatory and anti-fibrotic actions of colchicine may limit the progression of FSGS. To our surprise, no studies have explored colchicine in FSGS yet.

There is a general reluctance to use colchicine even in conditions with relatively established indications. This reluctance may be multifactorial in origin. One reason is the lack of sufficient human data except for the data that are available on the prevention of renal AA amyloidosis. Furthermore, there is no randomized controlled study conducted in other nephropathies. Another, perhaps more important, reason for the underutilization of colchicine is the concern due to drug-related toxicity. The literature is abounding with dramatic reports of adverse events related to colchicine use. In this regard, CKD patients may be at higher risk of being affected by colchicine toxicity. Thus, despite extensive liver metabolism [87,] urinary excretion via both glomerular filtration and tubular secretion clears up to 20% of the drug [88]. Creatinine clearance of <25 mL/min portends a high risk of colchicine accumulation [1]. However, there is controversy and inconsistency regarding colchicine dose adjustment in CKD. All authors recommend significant dose reductions in patients with glomerular filtration rate (GFR) <50 mL/min/1.73 m2. Some authors recommend complete avoidance of colchicine in patients with GFR <10 mL/min/1.73 m2 and patients undergoing hemodialysis [6,89]. However, these recommendations are not based on firm data. We recently studied the potential toxicity of colchicine under routine clinical practice conditions in 22 patients receiving long-term maintenance hemodialysis (>6 months, mean duration 8.9 ± 8.2 years) colchicine and 20 matched hemodialysis controls [90]. Four of 22 patients were using 0.5 mg/day, 4 patients were using 1.5 mg/day, and 14 patients were using 1 mg/day colchicine. There was no difference between the groups in terms of myo-neuropathic signs and symptoms, creatinine kinase and myoglobin levels, or blood counts except for white blood cell count, which was significantly higher in patients on colchicine treatment. Since this was not a randomized controlled study, bias cannot be excluded with certainty though. However, the study demonstrated that colchicine can be prescribed safely for those with advanced CKD for long periods of time.

Most of the reported toxicities in the literature had one or more “facilitating factors”, such as renal and/or hepatic dysfunction [91] or concomitant use of other drug(s) [92,93,94] affecting metabolism of colchicine. Drugs that increase the risk of colchicine toxicity via dual modulation of ABCB1 (P-glycoprotein) and CYP3A4 include the macrolide antibiotics erythromycin and clarithromycin and statins, among many others [6]. Cyclosporin and tacrolimus may also modulate P-glycoprotein.

Colchicine toxicity is dose-dependent and characterized by 3 sequential and overlapping phases [95]. The first phase reflects gastrointestinal mucosal damage and is characterized by nausea, vomiting, and diarrhea. Symptoms quickly resolve upon dose reduction or drug discontinuation. The second phase is characterized by multi-organ dysfunction and metabolic derangements. Almost every organ system may be affected. Myopathy, neuropathy, and bone-marrow suppression are frequent [96,97,98,99]. Practically it is not possible to measure blood colchicine levels in every laboratory. The third phase is recovery.

Taken together, colchicine toxicity follows a predictable sequence of events and related symptoms and is potentially reversible when recognized early. When initiated with low doses and up-titrated according to clinical response along with close surveillance in informed patients, colchicine can be used safely even in patients receiving hemodialysis. The United States Physicians' Desk Reference® recommends the reduction of colchicine dose with estimated GFR (eGFR) between 30 and 50 mL/min/1.73 m2 and 0.3 mg/day with clinical vigilance when eGFR is below 30 mL/min/1.73 m2[100]. The American College of Physicians' Drug Prescribing in Renal Failure booklet recommends 50-100% dose reduction with eGFR between 10 and 50 mL/min/1.73 m2, whereas 25% of usual dose with eGFR <10 mL/min/1.73 m2[101]. On the other hand, British National Formulary 61 recommends avoidance of colchicine in patients with eGFR <10 mL/min/1.73 m2[102].

Colchicine has a broad range of anti-inflammatory and antifibrotic properties. In addition to its time-honored anti-amyloidotic effects, experimental and clinical data have disclosed anti-fibrotic effects both in kidney diseases and several other fibrotic disorders. Recent preclinical evidence of benefit in DN and renal mass reduction along with evidence for biological plausibility given the current understanding of the pathophysiologic pathways leading to kidney disease support the exploration of the role of colchicine in clinical studies. In this regard, randomized clinical trials are ongoing for DN. Decades of clinical experience and an ever-increasing range of indications make it likely that we will continue to see colchicine around for a long time.

All authors approved the final version of manuscript.

There is no conflict of interest between authors.

1.
Nuki G: Colchicine: its mechanism of action and efficacy in crystal-induced inflammation. Curr Rheumatol Rep 2008;10:218-227.
2.
Leung YY, Yao Hui LL, Kraus VB: Colchicine - update on mechanisms of action and therapeutic uses. Semin Arthritis Rheum 2015;45:341-350.
3.
Verma S, Eikelboom JW, Nidorf SM, Al-Omran M, Gupta N, Teoh H, Friedrich JO: Colchicine in cardiac disease: a systematic review and meta-analysis of randomized controlled trials. BMC Cardiovasc Disord 2015;15:96.
4.
Bhattacharyya B, Panda D, Gupta S, Banerjee M: Anti-mitotic activity of colchicine and the structural basis for its interaction with tubulin. Med Res Rev 2008;28:155-183.
5.
Liu Y: Cellular and molecular mechanisms of renal fibrosis. Nat Rev Nephrol 2011;7:684-696.
6.
Terkeltaub RA: Colchicine update: 2008. Semin Arthritis Rheum 2009;38:411-419.
7.
Cronstein BN, Molad Y, Reibman J, Balakhane E, Levin RI, Weissmann G: Colchicine alters the quantitative and qualitative display of selectins on endothelial cells and neutrophils. J Clin Invest 1995;96:994-1002.
8.
Caner JE: Colchicine inhibition of chemotaxis. Arthritis Rheum 1965;8:757-764.
9.
Roberge CJ, Gaudry M, de Medicis R, Lussier A, Poubelle PE, Naccache PH: Crystal-induced neutrophil activation. IV. Specific inhibition of tyrosine phosphorylation by colchicine. J Clin Invest 1993;92:1722-1729.
10.
Martinon F, Petrilli V, Mayor A, Tardivel A, Tschopp J: Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature 2006;440:237-241.
11.
Viktorov AV, Yurkiv VA: Albendazole and colchicine modulate LPS-induced secretion of inflammatory mediators by liver macrophages. Bull Exp Biol Med 2011;151:683-685.
12.
Gagne V, Marois L, Levesque JM, Galarneau H, Lahoud MH, Caminschi I, Naccache PH, Tessier P, Fernandes MJ: Modulation of monosodium urate crystal-induced responses in neutrophils by the myeloid inhibitory C-type lectin-like receptor: potential therapeutic implications. Arthritis Res Ther 2013;15:R73.
13.
Niel E, Scherrmann JM: Colchicine today. Joint Bone Spine 2006;73:672-678.
14.
Quaggin SE, Kapus A: Scar wars: mapping the fate of epithelial-mesenchymal-myofibroblast transition. Kidney Int 2011;80:41-50.
15.
Bottinger EP: TGF-beta in renal injury and disease. Semin Nephrol 2007;27:309-320.
16.
Hinz B: Tissue stiffness, latent TGF-beta1 activation, and mechanical signal transduction: implications for the pathogenesis and treatment of fibrosis. Curr Rheumatol Rep 2009;11:120-126.
17.
Almasio PL, Floreani A, Chiaramonte M, Provenzano G, Battezzati P, Crosignani A, Podda M, Todros L, Rosina F, Saccoccio G, Manenti F, Ballardini G, Bianchi FP, Scheuer PJ, Davies SE, Craxi A: Multicentre randomized placebo-controlled trial of ursodeoxycholic acid with or without colchicine in symptomatic primary biliary cirrhosis. Aliment Pharmacol Ther 2000;14:1645-1652.
18.
Gong Y, Gluud C: Colchicine for primary biliary cirrhosis: a Cochrane Hepato-Biliary Group systematic review of randomized clinical trials. Am J Gastroenterol 2005;100:1876-1885.
19.
Lebleu VS, Sugimoto H, Miller CA, Gattone VH 2nd, Kalluri R: Lymphocytes are dispensable for glomerulonephritis but required for renal interstitial fibrosis in matrix defect-induced Alport renal disease. Lab Invest 2008;88:284-292.
20.
Nishida M, Hamaoka K: Macrophage phenotype and renal fibrosis in obstructive nephropathy. Nephron Exp Nephrol 2008;110:e31-e36.
21.
Ostermann D, Perico N, Imberti O, Barbui C, Bontempelli M, Remuzzi G: Colchicine allows prolonged survival of highly reactive renal allograft in the rat. J Am Soc Nephrol 1993;4:1294-1299.
22.
Perico N, Ostermann D, Bontempeill M, Morigi M, Amuchastegui CS, Zoja C, Akalin E, Sayegh MH, Remuzzi G: Colchicine interferes with L-selectin and leukocyte function-associated antigen-1 expression on human T lymphocytes and inhibits T cell activation. J Am Soc Nephrol 1996;7:594-601.
23.
Li C, Yang CW, Ahn HJ, Kim WY, Park CW, Park JH, Cha JH, Kim J, Kim YS, Bang BK: Colchicine suppresses osteopontin expression and inflammatory cell infiltration in chronic cyclosporine nephrotoxicity. Nephron 2002;92:422-430.
24.
Li JJ, Lee SH, Kim DK, Jin R, Jung DS, Kwak SJ, Kim SH, Han SH, Lee JE, Moon SJ, Ryu DR, Yoo TH, Han DS, Kang SW: Colchicine attenuates inflammatory cell infiltration and extracellular matrix accumulation in diabetic nephropathy. Am J Physiol Renal Physiol 2009;297:F200-F209.
25.
Vasyutina E, Treier M: Molecular mechanisms in renal degenerative disease. Semin Cell Dev Biol 2010;21:831-837.
26.
Liu Y: Renal fibrosis: new insights into the pathogenesis and therapeutics. Kidney Int 2006;69:213-217.
27.
Kopp JB, Factor VM, Mozes M, Nagy P, Sanderson N, Bottinger EP, Klotman PE, Thorgeirsson SS: Transgenic mice with increased plasma levels of TGF-beta 1 develop progressive renal disease. Lab Invest 1996;74:991-1003.
28.
Huang WY, Sun H, Pan XQ, Fei L, Guo M, Bao HY, Chen RH, Jiang XY: [Effects of colchicine on synthesis and excretion of cytokines and extracellular matrix by human renal fibroblasts]. Zhonghua Er Ke Za Zhi 2004;42:524-528.
29.
Diegelmann RF, Peterkofsky B: Inhibition of collagen secretion from bone and cultured fibroblasts by microtubular disruptive drugs. Proc Natl Acad Sci U S A 1972;69:892-896.
30.
Chung KY, Kang DS: Regulation of type I collagen and interstitial collagenase mRNA expression in human dermal fibroblasts by colchicine and D-penicillamine. Yonsei Med J 1999;40:490-495.
31.
Dehm P, Prockop DJ: Time lag in the secretion of collagen by matrix-free tendon cells and inhibition of the secretory process by colchicine and vinblastine. Biochim Biophys Acta 1972;264:375-382.
32.
Harris ED Jr, Krane SM: Effects of colchicine on collagenase in cultures of rheumatoid synovium. Arthritis Rheum 1971;14:669-684.
33.
Fell HB, Lawrence CE, Bagga MR, Hembry RM, Reynolds JJ: The degradation of collagen in pig synovium in vitro and the effect of colchicine. Matrix 1989;9:116-126.
34.
Galichon P, Hertig A: Epithelial to mesenchymal transition as a biomarker in renal fibrosis: are we ready for the bedside? Fibrogenesis Tissue Repair 2011;4:11.
35.
Humphreys BD, Lin SL, Kobayashi A, Hudson TE, Nowlin BT, Bonventre JV, Valerius MT, McMahon AP, Duffield JS: Fate tracing reveals the pericyte and not epithelial origin of myofibroblasts in kidney fibrosis. Am J Pathol 2010;176:85-97.
36.
Zeisberg EM, Potenta SE, Sugimoto H, Zeisberg M, Kalluri R: Fibroblasts in kidney fibrosis emerge via endothelial-to-mesenchymal transition. J Am Soc Nephrol 2008;19:2282-2287.
37.
Stafford SJ, Schwimer J, Anthony CT, Thomson JL, Wang YZ, Woltering EA: Colchicine and 2-methoxyestradiol inhibit human angiogenesis. J Surg Res 2005;125:104-108.
38.
Mabeta P, Pepper MS: A comparative study on the anti-angiogenic effects of DNA-damaging and cytoskeletal-disrupting agents. Angiogenesis 2009;12:81-90.
39.
Jung HI, Shin I, Park YM, Kang KW, Ha KS: Colchicine activates actin polymerization by microtubule depolymerization. Mol Cells 1997;7:431-437.
40.
Ozdemir BH, Ozdemir FN, Sezer S, Sar A, Haberal M: Does colchicine have an antifibrotic effect on development of interstitial fibrosis in renal allografts of recipients with familial Mediterranean fever? Transplant Proc 2006;38:473-476.
41.
Williams ME: Diabetic CKD/ESRD 2010: a progress report? Semin Dial 2010;23:129-133.
42.
Gross JL, de Azevedo MJ, Silveiro SP, Canani LH, Caramori ML, Zelmanovitz T: Diabetic nephropathy: diagnosis, prevention, and treatment. Diabetes Care 2005;28:164-176.
43.
Tesch GH: Macrophages and diabetic nephropathy. Semin Nephrol 2010;30:290-301.
44.
Chow F, Ozols E, Nikolic-Paterson DJ, Atkins RC, Tesch GH: Macrophages in mouse type 2 diabetic nephropathy: correlation with diabetic state and progressive renal injury. Kidney Int 2004;65:116-128.
45.
Sugimoto H, Shikata K, Hirata K, Akiyama K, Matsuda M, Kushiro M, Shikata Y, Miyatake N, Miyasaka M, Makino H: Increased expression of intercellular adhesion molecule-1 (ICAM-1) in diabetic rat glomeruli: glomerular hyperfiltration is a potential mechanism of ICAM-1 upregulation. Diabetes 1997;46:2075-2081.
46.
Coimbra TM, Janssen U, Grone HJ, Ostendorf T, Kunter U, Schmidt H, Brabant G, Floege J: Early events leading to renal injury in obese Zucker (fatty) rats with type II diabetes. Kidney Int 2000;57:167-182.
47.
Okada S, Shikata K, Matsuda M, Ogawa D, Usui H, Kido Y, Nagase R, Wada J, Shikata Y, Makino H: Intercellular adhesion molecule-1-deficient mice are resistant against renal injury after induction of diabetes. Diabetes 2003;52:2586-2593.
48.
Chow FY, Nikolic-Paterson DJ, Ozols E, Atkins RC, Tesch GH: Intercellular adhesion molecule-1 deficiency is protective against nephropathy in type 2 diabetic db/db mice. J Am Soc Nephrol 2005;16:1711-1722.
49.
Galkina E, Ley K: Leukocyte recruitment and vascular injury in diabetic nephropathy. J Am Soc Nephrol 2006;17:368-377.
50.
Takahashi T, Hato F, Yamane T, Inaba M, Okuno Y, Nishizawa Y, Kitagawa S: Increased spontaneous adherence of neutrophils from type 2 diabetic patients with overt proteinuria: possible role of the progression of diabetic nephropathy. Diabetes Care 2000;23:417-418.
51.
Watanabe A, Tomino Y, Yokoyama K, Koide H: Production of hydrogen peroxide by neutrophilic polymorphonuclear leukocytes in patients with diabetic nephropathy. J Clin Lab Anal 1993;7:209-213.
52.
Wierusz-Wysocka B, Wysocki H, Siekierka H, Wykretowicz A, Szczepanik A, Klimas R: Evidence of polymorphonuclear neutrophils (PMN) activation in patients with insulin-dependent diabetes mellitus. J Leukoc Biol 1987;42:519-523.
53.
van Oostrom AJ, van Wijk JP, Sijmonsma TP, Rabelink TJ, Castro Cabezas M: Increased expression of activation markers on monocytes and neutrophils in type 2 diabetes. Neth J Med 2004;62:320-325.
54.
Hatanaka E, Monteagudo PT, Marrocos MS, Campa A: Neutrophils and monocytes as potentially important sources of proinflammatory cytokines in diabetes. Clin Exp Immunol 2006;146:443-447.
55.
Guan T, Gao B, Chen G, Chen X, Janssen M, Uttarwar L, Ingram AJ, Krepinsky JC: Colchicine attenuates renal injury in a model of hypertensive chronic kidney disease. Am J Physiol Renal Physiol 2013;305:F1466-F1476.
56.
Honeck H, Gross V, Erdmann B, Kargel E, Neunaber R, Milia AF, Schneider W, Luft FC, Schunck WH: Cytochrome P450-dependent renal arachidonic acid metabolism in desoxycorticosterone acetate-salt hypertensive mice. Hypertension 2000;36:610-616.
57.
Nishikimi T, Matsuoka H: Molecular mechanisms and therapeutic strategies of chronic renal injury: renoprotective effect of rho-kinase inhibitor in hypertensive glomerulosclerosis. J Pharmacol Sci 2006;100:22-28.
58.
Akalin E, Hancock WW, Perico N, Remuzzi G, Imberti O, Carpenter CB, Sayegh MH: Blocking cell microtubule assembly inhibits the alloimmune response in vitro and prolongs renal allograft survival by inhibition of Th1 and sparing of Th2 cell function in vivo. J Am Soc Nephrol 1995;5:1418-1425.
59.
Mekori YA, Chowers Y, Ducker I, Klajman A: Inhibition of delayed hypersensitivity reactions by colchicine. II. Colchicine inhibits interferon-gamma induced expression of HLA-DR on gut epithelial cell line. Clin Exp Immunol 1989;78:230-232.
60.
Allen JN, Herzyk DJ, Wewers MD: Colchicine has opposite effects on interleukin-1 beta and tumor necrosis factor-alpha production. Am J Physiol 1991;261(4 pt 1):L315-L321.
61.
Freed BM, Lempert N, Lawrence DA: The inhibitory effects of N-ethylmaleimide, colchicine and cytochalasins on human T-cell functions. Int J Immunopharmacol 1989;11:459-465.
62.
Racusen LC, Regele H: The pathology of chronic allograft dysfunction. Kidney Int Suppl 2010;119:S27-S32.
63.
Menon MC, Murphy B: Maintenance immunosuppression in renal transplantation. Curr Opin Pharmacol 2013;13:662-671.
64.
Heemann U, Lutz J: Pathophysiology and treatment options of chronic renal allograft damage. Nephrol Dial Transplant 2013;28:2438-2446.
65.
Disel U, Paydas S, Dogan A, Gulfiliz G, Yavuz S: Effect of colchicine on cyclosporine nephrotoxicity, reduction of TGF-beta overexpression, apoptosis, and oxidative damage: an experimental animal study. Transplant Proc 2004;36:1372-1376.
66.
Sabry A, El-Husseini A, Sheashaa H, Abdel-Shafy E, El-Dahshan K, Abdel-Rahim M, Abdel-Kaleek E, Abo-Zena H: Colchicine vs. omega-3 fatty acids for prevention of chronic cyclosporine nephrotoxicity in Sprague Dawley rats: an experimental animal model. Arch Med Res 2006;37:933-940.
67.
Kosmadakis GCh, Papakonstantinou S, Theodoros C, Emmanouel P, Demetrios V, Nicolas Z: Characteristics of uremic pruritus in hemodialysis patients: data from a single center. Kidney Int 2008;74:962; author reply 962-963.
68.
Dember LM: Amyloidosis-associated kidney disease. J Am Soc Nephrol 2006;17:3458-3471.
69.
Herrera GA, Teng J, Turbat-Herrera EA: Renal amyloidosis: current views on pathogenesis and impact on diagnosis. Contrib Nephrol 2011;169:232-246.
70.
Glenner GG: Amyloid deposits and amyloidosis: the beta-fibrilloses (second of two parts). N Engl J Med 1980;302:1333-1343.
71.
Dispenzieri A, Kyle RA, Lacy MQ, Therneau TM, Larson DR, Plevak MF, Rajkumar SV, Fonseca R, Greipp PR, Witzig TE, Lust JA, Zeldenrust SR, Snow DS, Hayman SR, Litzow MR, Gastineau DA, Tefferi A, Inwards DJ, Micallef IN, Ansell SM, Porrata LF, Elliott MA, Gertz MA: Superior survival in primary systemic amyloidosis patients undergoing peripheral blood stem cell transplantation: a case-control study. Blood 2004;103:3960-3963.
72.
Kyle RA, Greipp PR, Garton JP, Gertz MA: Primary systemic amyloidosis. Comparison of melphalan/prednisone versus colchicine. Am J Med 1985;79:708-716.
73.
Cohen AS, Rubinow A, Anderson JJ, Skinner M, Mason JH, Libbey C, Kayne H: Survival of patients with primary (AL) amyloidosis. Colchicine-treated cases from 1976 to 1983 compared with cases seen in previous years (1961 to 1973). Am J Med 1987;82:1182-1190.
74.
Benson MD: Treatment of AL amyloidosis with melphalan, prednisone, and colchicine. Arthritis Rheum 1986;29:683-687.
75.
Skinner M, Anderson J, Simms R, Falk R, Wang M, Libbey C, Jones LA, Cohen AS: Treatment of 100 patients with primary amyloidosis: a randomized trial of melphalan, prednisone, and colchicine versus colchicine only. Am J Med 1996;100:290-298.
76.
Kyle RA, Gertz MA, Greipp PR, Witzig TE, Lust JA, Lacy MQ, Therneau TM: A trial of three regimens for primary amyloidosis: colchicine alone, melphalan and prednisone, and melphalan, prednisone, and colchicine. N Engl J Med 1997;336:1202-1207.
77.
Khalighi MA, Dean Wallace W, Palma-Diaz MF: Amyloid nephropathy. Clin Kidney J 2014;7:97-106.
78.
Livneh A, Zemer D, Langevitz P, Laor A, Sohar E, Pras M: Colchicine treatment of AA amyloidosis of familial Mediterranean fever. An analysis of factors affecting outcome. Arthritis Rheum 1994;37:1804-1811.
79.
Meneses CF, Egues CA, Uriarte M, Belzunegui J, Rezola M: Colchicine use in isolated renal AA amyloidosis. Reumatol Clin 2015;11:242-243.
80.
Livneh A, Zemer D, Siegal B, Laor A, Sohar E, Pras M: Colchicine prevents kidney transplant amyloidosis in familial Mediterranean fever. Nephron 1992;60:418-422.
81.
Abedi AS, Nakhjavani JM, Etemadi J: Long-term outcome of renal transplantation in patients with familial Mediterranean fever amyloidosis: a single-center experience. Transplant Proc 2013;45:3502-3504.
82.
Ben-Zvi I, Danilesko I, Yahalom G, Kukuy O, Rahamimov R, Livneh A, Kivity S: Risk factors for amyloidosis and impact of kidney transplantation on the course of familial Mediterranean fever. Isr Med Assoc J 2012;14:221-224.
83.
Erdem E, Karatas A, Kaya C, Dilek M, Yakupoglu YK, Arık N, Akpolat T: Renal transplantation in patients with familial Mediterranean fever. Clin Rheumatol 2012;31:1183-1186.
84.
Unverdi S, Inal S, Ceri M, Unverdi H, Batgi H, Tuna R, Ozturk MA, Guz G, Duranay M: Is colchicine therapy effective in all patients with secondary amyloidosis? Ren Fail 2013;35:1071-1074.
85.
Solak Y, Atalay H, Polat I, Biyik Z: Colchicine treatment in autosomal dominant polycystic kidney disease: many points in common. Med Hypotheses 2010;74:314-317.
86.
Malaga-Dieguez L, Bouhassira D, Gipson D, Trachtman H: Novel therapies for FSGS: preclinical and clinical studies. Adv Chronic Kidney Dis 2015;22:e1-e6.
87.
Leighton JA, Bay MK, Maldonado AL, Johnson RF, Schenker S, Speeg KV: The effect of liver dysfunction on colchicine pharmacokinetics in the rat. Hepatology 1990;11:210-215.
88.
de Lannoy IA, Mandin RS, Silverman M: Renal secretion of vinblastine, vincristine and colchicine in vivo. J Pharmacol Exp Ther 1994;268:388-395.
89.
Bhat A, Naguwa SM, Cheema GS, Gershwin ME: Colchicine revisited. Ann N Y Acad Sci 2009;1173:766-773.
90.
Solak Y, Atalay H, Biyik Z, Alibasic H, Gaipov A, Guney F, Kucuk A, Tonbul HZ, Yeksan M, Turk S: Colchicine toxicity in end-stage renal disease patients: a case-control study. Am J Ther 2014;21:e189-e195.
91.
Montseny JJ, Meyrier A, Gherardi RK: Colchicine toxicity in patients with chronic renal failure. Nephrol Dial Transplant 1996;11:2055-2058.
92.
Montiel V, Huberlant V, Vincent MF, Bonbled F, Hantson P: Multiple organ failure after an overdose of less than 0.4 mg/kg of colchicine: role of coingestants and drugs during intensive care management. Clin Toxicol (Phila) 2010;48:845-848.
93.
Lonesky TA, Kreuter JD, Wortmann RL, Rhodes CH: Hydroxychloroquine and colchicine induced myopathy. J Rheumatol 2009;36:2617-2618.
94.
Bouquie R, Deslandes G, Renaud C, Dailly E, Haloun A, Jolliet P: Colchicine-induced rhabdomyolysis in a heart/lung transplant patient with concurrent use of cyclosporin, pravastatin, and azithromycin. J Clin Rheumatol 2011;17:28-30.
95.
Finkelstein Y, Aks SE, Hutson JR, Juurlink DN, Nguyen P, Dubnov-Raz G, Pollak U, Koren G, Bentur Y: Colchicine poisoning: the dark side of an ancient drug. Clin Toxicol (Phila) 2010;48:407-414.
96.
Dickinson M, Juneja S: Haematological toxicity of colchicine. Br J Haematol 2009;146:465.
97.
van der Velden W, Huussen J, Ter Laak H, de Sevaux R: Colchicine-induced neuromyopathy in a patient with chronic renal failure: the role of clarithromycin. Neth J Med 2008;66:204-206.
98.
Pirzada NA, Medell M, Ali II: Colchicine induced neuromyopathy in a patient with normal renal function. J Clin Rheumatol 2001;7:374-376.
99.
Older SA, Finbloom DS, Pezeshkpour GH: Colchicine myoneuropathy and renal dysfunction. Ann Rheum Dis 1992;51:1343-1344.
100.
Physicians' Desk Reference (PDR). 2011. http://www.Pdr.Net/drugpages/concisemonograph.Aspx?Concise=3048 (accessed February 18, 2012).
101.
Brier ME, Aronoff GR: Drug Prescribing in Renal Failure, ed 5. Philadelphia, American College of Physicians, 2007.
102.
Joint Formulary Committee: British National Formulary, ed 48. London, British Medical Association and Royal Pharmaceutical Society of Great Britain, 2004.
103.
Sagiroglu T, Sayhan MB, Yagci MA, Yalta T, Sagiroglu G, Copuroglu E, Oguz S: Comparison of sirolimus and colchicine treatment on the development of peritoneal fibrozis in rats having peritoneal dialysis. Balkan Med J 2015;32:101-106.
104.
Bozkurt D, Bicak S, Sipahi S, Taskin H, Hur E, Ertilav M, Sen S, Duman S: The effects of colchicine on the progression and regression of encapsulating peritoneal sclerosis. Perit Dial Int 2008;28(suppl 5):S53-S57.
105.
Sayarlioglu H, Dogan E, Erkoc R, Ozbek H, Bayram I, Sayarlioglu M, Sekeroglu R, Bozkurt H: The effect of colchicine on the peritoneal membrane. Ren Fail 2006;28:69-75.
106.
Chen W, Huang SL, Huang CS, Tsai MC, Lai HM, Lui CC, Eng HL, Chang HW, Lee CH, Chuang FR: Nephrogenic systemic fibrosis in advanced chronic kidney disease: a single hospital's experience in Taiwan. Eur J Dermatol 2009;19:44-49.
107.
de Socio G, Verrecchia E, Fonnesu C, Giovinale M, Gasbarrini GB, Manna R: Effectiveness of colchicine therapy in 4 cases of retroperitoneal fibrosis associated with autoinflammatory diseases. J Rheumatol 2010;37:1971-1972.
108.
Vega J, Goecke H, Tapia H, Labarca E, Santamarina M, Martinez G: Treatment of idiopathic retroperitoneal fibrosis with colchicine and steroids: a case series. Am J Kidney Dis 2009;53:628-637.
109.
Li C, Yang CW, Ahn HJ, Kim WY, Park CW, Park JH, Lee MJ, Yang JH, Kim YS, Bang BK: Colchicine decreases apoptotic cell death in chronic cyclosporine nephrotoxicity. J Lab Clin Med 2002;139:364-371.
110.
Sobh M, Sabry A, Moustafa F, Foda MA, Sally S, Ghoneim M: Effect of colchicine on chronic ciclosporin nephrotoxicity in Sprague-Dawley rats. Nephron 1998;79:452-457.
111.
Zhang G, el Nahas AM: Colchicine and the progression of experimental renal fibrosis. Nephrol Dial Transplant 1996;11:559-560.
112.
McClurkin C Jr, Phan SH, Hsu CH, Patel SR, Spicker JK, Kshirsagar AM, Yuan WY, Wiggins RC: Moderate protection of renal function and reduction of fibrosis by colchicine in a model of anti-GBM disease in the rabbit. J Am Soc Nephrol 1990;1:257-265.
Copyright / Drug Dosage / Disclaimer
Copyright: All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means, electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.