LFS-14202; No of Pages 10 Life Sciences xxx (2014) xxx–xxx

Contents lists available at ScienceDirect

Life Sciences

1

Review Article

2

Role of inflammation in the aging bone

3Q3

Samir M. Abdelmagid a, Mary F. Barbe c, Fayez F. Safadi a,b,⁎

4Q4 5Q6 6Q8

a

7

a r t i c l e

8 9 10 11

Article history: Received 6 June 2014 Accepted 17 November 2014 Available online xxxx

12 13 14 15 16 17

Keywords: Aging Inflammation Macrophages Osteoblasts Osteoclasts

O

R O

i n f o

a b s t r a c t

P

Chronic inflammation in aging is characterized by increased inflammatory cytokines, bone loss, decreased adaptation, and defective tissue repair in response to injury. Aging leads to inherent changes in mesenchymal stem cell (MSC) differentiation, resulting in impaired osteoblastogenesis. Also, the pro-inflammatory cytokines increase with aging, leading to enhanced myelopoiesis and osteoclastogenesis. Bone marrow macrophages (BMMs) play pivotal roles in osteoblast differentiation, the maintenance of hematopoietic stem cells (HSCs), and subsequent bone repair. However, during aging, little is known about the role of macrophages in the differentiation and function of MSC and HSC. Aged mammals have higher circulating pro-inflammatory cytokines than young adults, supporting the hypothesis of increased inflammation with aging. This review will aid on the understanding of the potential role of pro-inflammatory (M1) and anti-inflammatory (M2) macrophages in differentiation and function of osteoblasts and osteoclasts. © 2014 Published by Elsevier Inc.

D

c

Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED) School of Medicine, Rootstown, OH 44272, USA School of Biomedical Sciences, Kent State University, Kent, OH 44240, USA Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, PA 19140, USA

E

b

52 53 54 55

Q11 Q10

R

R

N C O

49 50 Q12 Introduction 51

. . . . . . . . . . . . . .

E

Introduction . . . . . . . . . . . . . . . . . . . . . . . Inflammation in aging . . . . . . . . . . . . . . . . . . . Macrophages and aging . . . . . . . . . . . . . . . . . . Macrophage polarization . . . . . . . . . . . . . . . . . . Cumulative trauma disorders . . . . . . . . . . . . . An operant animal model of repetitive strain injury . . . Repetitive strain injury and aging . . . . . . . . . . . Inflammation and bone homeostasis . . . . . . . . . . . . Osteoimmunology . . . . . . . . . . . . . . . . . . . . . Macrophages and tissue regeneration at the site of tissue injury Conclusions and future directions . . . . . . . . . . . Conflict of interest statement . . . . . . . . . . . . . . . . Uncited reference . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . .

C

Contents

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

U

35 36 37 38 39 40 41 42 43 44 45 46 47 48

18 19 20 21 22 23 24 25 26 27 28

T

32 30 29 31 34 33

F

journal homepage: www.elsevier.com/locate/lifescie

The population of Americans aged 65 and older is expected to double in the next 25 years due to increased life expectancy. The anticipated growth in the aging population will result in an expected 25% increase in health care costs by 2030 [54]. Aging is associated with chronic inflammation and with the consequent higher risk for diseases, morbidity ⁎ Corresponding author at: Department of Anatomy and Neurobiology, Northeast Ohio Medical University, 4209 State Rt. 44, Rootstown, OH 44224, USA. Tel.: +1 330 325 6596; fax: +1 330 325 5916. E-mail address: [email protected] (F.F. Safadi).

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

. . . . . . . . . . . . . .

0 0 0 0 0 0 0 0 0 0 0 0 0 0

and perhaps mortality. Chronic systemic inflammation is a common problem associated with aging and is responsible for 7 out of 10 deaths in the elderly, resulting in more than 1.7 million deaths every year and accounting for more than 75% of two trillion dollars spent on health care per year in the U.S. [64,166]. Serum levels of circulating proinflammatory cytokines, such as interleukin (IL)-6 and tumor necrosis factor (TNF-α) are typically elevated two-to four-fold in the elderly when compared to the young population, even in the absence of chronic disease [41]. These multifunctional cytokines have been associated with morbidity and mortality in the elderly. There is supportive evidence for a direct role of TNF-α in the pathogeneses of atherosclerosis, diabetes mellitus type-2, and Alzheimer's disease in aged individuals

http://dx.doi.org/10.1016/j.lfs.2014.11.011 0024-3205/© 2014 Published by Elsevier Inc.

Please cite this article as: S.M. Abdelmagid, et al., Role of inflammation in the aging bone..., Life Sci (2014), http://dx.doi.org/10.1016/ j.lfs.2014.11.011

56 57 58 59 60 61 62 63 64 65 66 67

C

E

111 112 113 114 115 116 117 118 119 120 121 122 123

Although, it is well studied that the adaptive immune system including both B and T lymphocytes deteriorate with advancing age [46,119], the effects of the innate immune response mediated by macrophages have been under-investigated. One aspect that has not been explored is the effect of aging on macrophage polarization. Macrophages isolated from aged humans and mice display reduced functions, ranging from a defective response in the early immune defense to decreased efficiency in the development of specific immune reactions [116,117,151,168]. Macrophages can be classified into classically activated M1 cells, or, alternatively, as activated M2 macrophages, based on their polarization status. M1 macrophages can be induced by lipopolysaccharide derived from bacteria (LPS) or a combination of T helper cell (Th1) cytokines, such as IFN-γ and TNF-α. Inflammatory M1 macrophages also up-regulate pro-inflammatory mediators, including IL-1β, TNF-α, IL-6, IL-12, and their receptors (Table 1). Moreover, M1 macrophages increase the production of reactive oxygen species (ROS), inducible nitric oxide synthase (iNOS) and other nitrogen intermediates [83,167]. In contrast, M2 macrophages can be induced by Th2 cytokines, such as M-CSF and IL-4 [82]. Anti-inflammatory M2 macrophages upregulate the expression of arginase-1 (Arg-1), scavenger and mannose

124

U

N

C

O

R

R

109 110

Macrophage polarization

T

Aging results from the accumulation of detrimental changes at the cellular and molecular levels in all organs and tissues, changes that ulti76 mately lead to increased risk for several diseases [164]. Young adult 77 mammals are able to survive their reproductive years because of strong 78 immune and inflammation responses. However, the same immune 79 mechanisms can lead to deleterious effects in humans that survive to 80 older ages [137]. Aging is a complex process in which body organs and 81 tissues lose their structural integrity leading to increased prevalence 82 for age-related diseases, such as osteoporosis, osteoarthritis, dementia 83 and cancer. Although the etiology of the aging process is not fully under84 stood [164], altered inflammatory processes play a major role in aging 85 [56]. 86 Inflammation occurs subsequent to trauma or infection at the cellu87 lar level [22,29,30]. As a result, inflammatory cells, e.g. macrophages and 88 monocytes, are activated and release several inflammatory cytokines 89 into the systemic circulation, including IL-1β, IL-6 and TNF-α. These cy90 tokines in return are responsible for the humoral immune responses 91 [28,29,52]. The aim of inflammation is to initiate the repair processes 92 that restore the tissue to its physiological condition [75] (Fig. 1). 93 Inflammatory cascade can also induce tissue catabolism if not regulated 94 properly. Several epidemiological studies have characterized the im95 mune response in aged individuals. Levels of inflammatory cytokines 96 and mediators increase with aging even in the absence of acute 97 infections or other physiologic stressors [164]. Chronic increases of in98 flammatory mediators underlie many aging-related conditions, such 99 Q13 as autoimmune diseases and malignancies [56,171]. For example, rheu100 matoid arthritis is characterized by high levels of pro-inflammatory cy101 tokines not only in the patient serum, but also in their affected joints 102 [81]. Similarly, in multiple myeloma, a neoplasm of B-cell origin [15], 103 the cancer cells secrete numerous pro-inflammatory cytokine-induced 104 chemotaxis of osteoclasts, resulting in profound bone resorption and 105 enhanced tumor growth [128,182].

107 108

F

74 75

Studies from other groups have shown that anti-inflammatory macrophages (M2) regulate MSC differentiation needed for tissue repair [120]. Based on our previously published reports and generated preliminary data, we suggest that macrophage polarization is shifted towards the pro-inflammatory macrophages (M1) as a consequence of aging. This phenotypic change is exacerbated with chronic inflammation or injury, due to the steady release of inflammatory cytokines from injured cells and tissues, compared with the acute increases of the same cytokines [181]. Combined, this results in reduced bone quality in aged populations (Fig. 2). However, the role of macrophage polarization in bone homeostasis has not been previously studied. We hypothesize that age, injury and chronic inflammation alter macrophage polarization in a manner that negatively influences the bone homeostasis. In this review, we propose to discuss the role of macrophage polarization in the recruitment and differentiation of MSC and HSC into osteoblasts and osteoclasts in young adults and aged mammals, respectively.

O

Inflammation in aging

106

R O

73

70 71

Macrophages and aging

P

72

[73,100,123]. High levels of circulating IL-6 may also be a risk factor for thromboembolic complications [131]. Furthermore, even in healthy, elderly populations, high circulating levels of inflammatory cytokines are predictive of mortality [41]. The objective of this review is to characterize the potential role(s) of macrophages in age-related bone loss.

D

68 69

S.M. Abdelmagid et al. / Life Sciences xxx (2014) xxx–xxx

E

2

Fig. 1. Proposed model for the effects of M1/M2 polarization on osteoblast differentiation. M1 macrophages (macs) decrease the differentiation of MSC into osteoblasts (OB) in young adults. However, with aging, M1 macs are inherently increased, leading to further reduction in MSC into OB. M2 macs are increased with inflammation in young adults, a change that will result in increased MSC differentiation and OB function. The latter will decrease with aging, due to reduced M2 cells.

Please cite this article as: S.M. Abdelmagid, et al., Role of inflammation in the aging bone..., Life Sci (2014), http://dx.doi.org/10.1016/ j.lfs.2014.11.011

125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143

3

R O

O

F

S.M. Abdelmagid et al. / Life Sciences xxx (2014) xxx–xxx

Cumulative trauma disorders

159 160

The term Work-related Musculoskeletal Disorders (WMSDs) represents a model of chronic inflammation, and encompasses neuromusculoskeletal disorders arising from repeated task performance with submaximal levels of physical exertion, or work in awkward postures [27]. WMSDs are a leading cause of long-term pain and physical disability world-wide [39,43,91,93,179], with diagnostic signs of tendinopathies, neuritis, myositis, bursitis, osteoarthritis and bone stress fractures [44,84]. In 2011, WMSDs accounted for 33% of all lost work time, workplace injuries and illnesses in the U.S. and a median of 11 days absence from work [43]. Work-related MSDs in the U.S. are associated with 130 million health care encounters and are estimated to cost over $50 billion annually [138]. In 2011, the number of occupational injuries involving days away from work due to hand and wrist injuries was 140,460 and 47,550, respectively, and their incidence rates were 13.9 and 4.7 per 10,00 workers [43]. Work-related MSDs often

161 162 163 164 165 166 167 168 169 170 171 172 173

C

E

R

154 155

R

153

N C O

151 152

U

149 150

t1:1 Q1

Table 1

t1:2

M1 markers

M2 markers

t1:3 t1:4 t1:5 t1:6 t1:7 t1:8

CD 68 (ED1) IL-1β (cytokine) IL-6 (cytokine) TNF-α (cytokine) CCL8 (chemokine) CXCL9 (chemokine)

CD163 (ED2) IL-10 (cytokine) TGF-β IL21R (cytokine-R) CCL18 (chemokine) CXCL1 (chemokine)

result from physical demands placed upon the musculoskeletal system in the workplace [36,139]. Several risk factors of occupationally related MSDs have been identified, including forceful exertions, repetitive motion, and non-neutral body postures [154]. Repetitive trauma, especially in aged individuals, results in structural damage of tissues and cell membranes. Part of the etiology of repeated trauma is that intracellular factors, including inflammatory cytokines, diffuse out through disrupted plasma membranes of damaged cells into the extracellular matrix [28,29,121]. These fragmented plasma membranes are chemotactic stimulants and induce lymphocyte and macrophage infiltration into the site of injury, as do inflammatory cytokines released from damaged cells. Tissue fibroblasts, satellite, endothelial cells and infiltrating macrophages also release cytokines in response to tissue damage [13,48,49,77,110]. The released cytokines further stimulate cell proliferation and macrophage infiltration, leading to edema, inflammation and pain [48], although some cytokines stimulate proliferation and differentiation of stem cells instead, which can lead to tissue regeneration [47,77,109,110]. Inflammatory cytokines released during the acute inflammatory phase, such as IL-1α, IL-1β and TNF-α, are known to stimulate proliferation and maturation of macrophages and other mononuclear cells [47,77,109,110] that in turn further activate macrophages and the production of additional inflammatory cytokines [13,14,88,136]. With regard to tissue catabolism, IL-1α, for example, serves as a potent stimulator of bone resorption [110,126]. Its increase can lead to activation of macrophages and osteoclasts in bone, enhancing their phagocytic function (and therefore bone resorption) as well as the release of additional cytokines. These events combined will initiate a vicious cycle of tissue damage and chronic inflammation, as described in detail previously [20,28,29,32]. In support of this, other findings have detected inflammation biomarkers in serum of patients with upper extremity WMSDs, including IL-6, TNFα, and members of the IL-1 family [50,158,160]. These increases correlated with symptom severity in patients with upper extremity WMSDs [50,160]. Also, increased serum levels of carboxyterminal collagen type I peptide (CTX-1), a serum biomarker of bone resorption, have been identified in workers involved in heavy manual lifting [105–107]. For example, increased CTX-1 has been associated with spinal shrinkage (assayed as stature loss) in healthy young nurses performing patient handling activities [105]. Since serum CTX-1 is released by activated osteoclasts, these studies suggest that bone resorptive events are occurring with WMSDs, although a causal relationship has yet to be established in this patient population.

D

158

147 148

T

156 157

receptors, as well as several intracellular proteins, such as Found in Inflammatory Zone 1 (FIZZ-1), T-lymphocyte-derived eosinophil chemotactic factor (ECF-L) and Ym-1. Arginine is metabolized by Arg-1 to ornithine and polyamines in M2 macrophages, thus, decreasing substrate availability for M1 macrophages to produce nitric oxide (NO). FIZZ-1 and Ym1 are produced in large amounts during allergic inflammation and other pathological states in which a highly polarized Th2 response is prevalent [130,132,133] (Fig. 3). It has been well reported that Th1 cells are increased with age, unlike Th2 cells, and that the ratio of Th1/Th2 increases with aging [162,173] (Fig. 4). The altered percentage of Th1 to Th2 cells with aging contributes to the polarization shift of macrophages towards the classically activated M1 phenotype, which in turn explains, at least in part, the chronic inflammatory events observed with aging [40,117,176] (Fig. 2).

145 146

E

144

P

Fig. 2. Proposed model for the effects of M1/M2 polarization on osteoclast differentiation. In contrast to MSC, M1 macs increase HSC differentiation and therefore osteoclast (OC) function. However, with aging, M1 macs further stimulate HSC differentiation with enhanced OC differentiation, resulting in bone loss. M2 macs are increased with inflammation in young adults, but to a lower extent with aging resulting in a reduced HSC differentiation into OC.

Please cite this article as: S.M. Abdelmagid, et al., Role of inflammation in the aging bone..., Life Sci (2014), http://dx.doi.org/10.1016/ j.lfs.2014.11.011

174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 Q14 190 191 192 193 194 Q15 195 196 Q16 197 198 199 200 201 202 203 204 205 206 207 Q17 208 209 210 211 212 Q18 213 214 215

S.M. Abdelmagid et al. / Life Sciences xxx (2014) xxx–xxx

T

E

D

P

R O

O

F

4

An operant animal model of repetitive strain injury

217

R

216

E

C

Fig. 3. Proposed model for M1/M2 polarization in young adults. M1 macs differentiate in response to INF-γ from Th1 cells. M2 macs differentiate in response to IL-4 from Th2 cells. Chronic inflammation in young adults, stimulates production of pro-inflammatory cytokines in M1 macs, followed by production of anti-inflammatory cytokines in M2 macs, with resultant net tissue adaptation (increased bone).

U

N

C

O

R

The laboratory of Dr. Mary Barbe has developed a unique repetitive 218 and forceful reaching rat model in which rats tolerantly perform 219 upper extremity repetitive reaching tasks [4,22,58,59,66,155,157]. The 220 reach rates and force levels used in this model were determined from 221 epidemiological and clinical studies as risks for WMSDs in humans 222 [11,19,21,23,24,26,28,31–33,51,52,58,60,69,71,72,156]. In this review, 223 we focus on the effects of performance of a high repetition low force 224 task as a model of chronic inflammation, it is as described next. Rats per225 form the HRLF task regimens for 2 h/day, 3 days/week for 12 weeks, 226 while daily task sessions are divided into 4, half-hour sessions separated 227 by 1.5 h in order to avoid satiation. For high repetition tasks, rats are 228 cued to reach at target rates of 4 reaches/min. Task rats reach forward 229 into a portal, extend their forearm, grab a lever and then exerted a target 230 isometric pull for at least 50 ms at a force effort of either 15% (0.23 N) of 231 their average maximum pulling force (±5%) for low force tasks. Signs 232 and symptoms indicative of the development of carpal tunnel syn233 drome, a known type of WMSD, have been observed in this model, 234 including forepaw hypersensitivity, declines in grip strength and reach 235 performance, as well as degraded myelin and decreased conduction 236 Q19 velocity in the median nerve at the level of the wrist [58,59,61,68–71, 237 96,97]. The sensorimotor declines correlated with increased tissue in238 flammation (macrophages and cytokines) and increased levels of 239 Q20 serum inflammatory cytokines [4,58,61,70,96]. Performance of repeti240 tive reaching also induced exposure-dependent changes in inflammato241 ry cytokines, osteoclast and osteoblast numbers in involved forelimb 242 bones [25,33,67,155,157]. Rats performing a low demand reaching

task for 12 weeks, had a transient increase in osteoclasts in forelimb bones in weeks 4–6 that resolved by week 12, a time point associated with bone adaptation [25,33]. In contrast, performance of high force tasks for 12 weeks resulted in prolonged increases in bone inflammatory cytokines, degradation in radiocarpal joints, metaphyseal cortical bone thinning, and increased serum biomarkers of bone and cartilage degradation (TRAP5b and C1,2C) [67,155,157]. Radio-carpal joint degradation was attenuated with oral ibuprofen treatment, indicating that joint degeneration is linked to inflammation in this model. Interestingly, aging increased the serum and tissue inflammatory cytokines and tissue macrophages in this WMSD rat model. In addition, there was also increased nerve and tendon degradative changes in aged rats performing a moderate demand task for 12 weeks, changes that correlated with increased tissue macrophages and inflammatory cytokines [70,96,181].

243

Repetitive strain injury and aging

257

As mentioned above, performance of upper extremity high repetition low force (HRLF) tasks for prolonged periods results in increased sera and tissue levels of pro-inflammatory cytokines and chemokines. This response was exacerbated with aging [70,96,181], supporting a hypothesis of both age- and repetitive task-induced increases in these cytokines. Dr. Barbe's group also observed a transient increase in serum proinflammatory cytokines mediated by M1 (pro-inflammatory) macrophages, such as, IFNγ, in young adult rats at week 6 of the HRLF task. However, this serum inflammatory cytokine response was resolved by week 12, coincident with increased serum IL-10, an M2- (anti-inflammatory) mediated cytokine. The latter is indicative of an M2 macrophage

258 259

Please cite this article as: S.M. Abdelmagid, et al., Role of inflammation in the aging bone..., Life Sci (2014), http://dx.doi.org/10.1016/ j.lfs.2014.11.011

244 245 Q21 246 247 248 249 Q22 250 251 252 253 254 255 256 Q23

260 261 Q24 262 263 264 265 266 267 268

5

T

E

D

P

R O

O

F

S.M. Abdelmagid et al. / Life Sciences xxx (2014) xxx–xxx

C

Fig. 4. Proposed model for M1/M2 polarization in aged mammals. In contrast to young adults, in aged mammals, M1 pro-inflammatory cytokines are inherently increased due to reversal of Th1/Th2 ratio; the tissue injury progressively increases M1 and decreases M2 responses, with a net loss of an adaptive response resulting in bone resorption and erosions. The combination of age and tissue injury shifts the mac polarization towards the M1 phenotype.

269

E

mediated resolution of the systemic cytokine response in young adult rats, despite continued task performance. This resolution was absent in aged rats that performed the 12-week HRLF, suggestive of a loss of tissue 272 Q25 repair or adaptation in the aged rats [70,96,181]. Although this loss of 273 tissue adaptation and repair with aging is apparent in the literature, the 274 underlying mechanisms are not fully understood.

R

R

270 271

Inflammation and bone homeostasis

276 277

Osteoporosis is a growing public health problem, in part because of the increasing numbers of people living beyond 65 years of age [127,169]. The National Osteoporosis Foundation estimates that there are 29.5 million women and 11.7 million men in the U.S. with osteoporosis. It is characterized by low bone mass, significant destruction of trabecular architecture and loss of connectivity between plates of trabecular bone, leading to increased bone fragility and fracture risk [127,169]. The estimated cost of treating patients hospitalized with a diagnosis of osteoporosis is $19.1 billion per year [65]. Eighty percent of patients with osteoporosis are women, largely due to the marked reduction in bone density associated with estrogen withdrawal during menopause. Bone resorption has been associated with a significant increase of pro-inflammatory cytokines in patients with inflammatory diseases. For example, chronic gingivitis; a periodontal disease is associated with increased pro-inflammatory cytokines and osteoclastmediated destruction of alveolar bone in the jaw. Also, rheumatoid arthritis is associated with increased pro-inflammatory cytokines in the affected joints, as well as osteoclast-mediated bone erosion on the margins of these joints [81]. A long list of pro-inflammatory cytokines is capable of stimulating osteoclastic bone resorption, including IL-1, TNF-α, IL-6, IL-11, IL-15 and IL-17 [34,103,125,178].

280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296

U

278 279

N C O

275

Osteoimmunology

297

Several bone diseases have been linked to the immune cell activity, such as the release of -pro-inflammatory cytokines with subsequent production of receptor activator of nuclear factor kappa-B ligand (RANKL) by osteoblasts [174]. RANKL stimulates osteoclast formation and bone erosions resulting in the development of osteolytic lesions in patients with rheumatoid arthritis [63]. The mechanism of rheumatoid arthritis-induced bone loss is explained by the infiltration of synovium and surrounding tissues of the affected joints with inflammatory cells, such as T lymphocytes and macrophages. T lymphocytes stimulate osteolysis, an event mediated by osteoclast differentiation and activation. Initial studies showed that activated T lymphocytes, or their culture supernatants, stimulate osteoclastogenesis in vitro [101]. Activated macrophages also secret pro-inflammatory cytokines such as TNF-α, IL-1 and IL-6 that have a synergistic effect on RANKL release by osteoblasts [8,9,86]. These inflammatory cytokines also direct osteoclast formation through a RANKL/RANK-independent mechanism, as previously reported [92,99,170]. In addition, activated macrophages and T lymphocytes produce macrophage migration inhibitory factor (MIF) [35], a chemokine that stimulates expression of matrix metalloproteinase (MMP)-9 and -13 by osteoblasts [143] and that supports RANKL-stimulated osteoclastogenesis in vitro [98]. Previous studies reported the osteoporotic phenotype with high bone turnover in mice overexpressing MIF [144], while mice lacking MIF do not exhibit bone loss [146]. Other devastating disease with high incidence in the elderly is multiple myeloma; a plasma B-cell tumor that arises in the bone marrow and quickly metastasizes to the skeleton. The tumor cells start and maintain a chronic inflammatory cascade and produce a chemokine called macrophage inflammatory protein (MIP)-1α and β. This

298

Please cite this article as: S.M. Abdelmagid, et al., Role of inflammation in the aging bone..., Life Sci (2014), http://dx.doi.org/10.1016/ j.lfs.2014.11.011

299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325

S.M. Abdelmagid et al. / Life Sciences xxx (2014) xxx–xxx

P

R O

O

F

not well understood. Our group has previously reported early tissue injury marked by a transient increase in serum and tissue inflammatory cytokines and a decrease in grip strength in young adults performing repetitive strain tasks [20,25,33]. Recent studies showed an induction of M1 polarization and a persistent increase of M1 cytokines (IFN-γ) in aged versus young adults indicating a chronic inflammation. This response was even greater in aged versus young adults performing the repetitive strain task [70,96,180]. We have preliminary data showing that serum from aged HRLF rats (model of repetitive strain injury) inhibits MSC proliferation, compared to serum from young HRLF rats. In addition, we have preliminary evidence that serum from aged HRLF rats increased HSC proliferation, compared to serum from young HRLF rats. These findings suggest that serum anti-inflammatory cytokines in young rats are preferentially contributed by alternatively activated M2 macrophages that likely have a reparative role mediated by MSC proliferation and then differentiation into osteoblasts at the site of injury, a response that should lead to adaptive bone formation. Conversely, serum cytokines in aged rats that are preferentially contributed by classically activated M1 macrophages have a pathological role in mediating HSC proliferation and differentiation into osteoclasts, a response that should lead to enhanced bone resorption. Studies confirming these hypotheses are currently underway in our laboratories.

C

E

R

R

O

N

C

377 378

Macrophages and tissue regeneration at the site of tissue injury

U

376

The site of a tissue injury is largely associated with a dense population of infiltrated macrophages with heterogeneous phenotypes and 379 an inflammatory response that is mediated by these cells [12]. Thus, 380 macrophages modulate the microenvironment at the site of injuries 381 Q28 [12]. Macrophage polarization (M1 versus M2) differentially regulates 382 MSC and HSC differentiation in bone tissue. This regulatory mechanism 383 mediates either a regenerative or pathological response based on M1/ 384 M2 shift mediating MSC and HSC differentiation, respectively [55,74]. 385 Recent studies show that macrophage polarization regulates certain 386 physiological, metabolic conditions and regenerative functions [37,38]. 387 The nature of the complex interplay between the M1 and M2 macro388 phages and how these interactions influence the ability of MSC389 mediated tissue regeneration or HSC-mediated tissue pathology are

390 391 392 393 394 395 396 397 Q29 398 399 400 401 402 403 404 405 406 407 408 409 410 411

Conclusions and future directions

412

The role of inflammatory macrophages in age-associated bone loss is under investigated. Elucidation of the complex interplay between chronic inflammation and altered bone homeostasis is a subject of intense clinical interest. Treatment of aged patients with generalized bone loss, such as osteoporosis and/or chronic inflammation is a health challenge. Since bone homeostasis in aged patients is likely altered due to chronic inflammation, future studies should focus on new strategies to reduce age-related inflammation for improved quality of life in the elderly. This information will help in developing new therapeutic strategies to selectively enhance bone formation in aged patients suffering from chronic inflammation and significant bone loss. For example, stem cell therapy is a hot topic for treatment of several other pathological bone diseases. One suggested intervention is the treatment of aged osteoporotic patients with bone marrow derived-mesenchymal stem cells and/or anti-inflammatory repair macrophages isolated from their young siblings [118]. We also need to improve our understanding of the regulatory mechanisms of inflammation in skeletal tissues of aged mammals at rest and following injury that will help develop novel and specific anti-inflammatory medications. In addition, we need to explore the advantages of dietary modifications and regular exercise training to subside inflammation in skeletal tissues of aged mammals. Moderate exercise, in particular resistance training, is effective in maintaining the skeletal mass in aged mammals [102,149]. Moreover, moderate exercise is also effective in decreasing systemic inflammation [42,62]. Reducing inflammation in skeletal tissues will help to restrict muscle atrophy and bone loss, enhance muscular strength, and reduce the incidence of osteoporosis and fracture risk in aged mammals [78,89,124]. More research is required to establish the potential mechanisms of macrophages and inflammatory cytokines in regulating bone homeostasis and that will be critical over the course of the next 30 years, during which the world's aged population is expected to grow exponentially.

413

Conflict of interest statement

444 Q31

T

chemokine not only triggers proliferation and survival of tumor cells, but also enhances osteoclast formation-mediated bone resorption 328 resulting in osteolytic lesions and leading to increased morbidity and 329 mortality [7,87,112]. 330 In MSC and osteoblasts, numerous cytokines are reported to regulate 331 their proliferation, survival and differentiation. TNF-α is a pro-apoptotic 332 for osteoblasts [94], possibly through induction of Fas–FasL interaction 333 [95]. TNF-α is also reported to inhibit osteoblast differentiation [79,80, 334 108,135]. IFN-γ, IL-1 and TNF-α inhibit collagen synthesis by osteo335 blasts [45,53,165]. Several anti-inflammatory cytokines counteract the 336 effects of pro-inflammatory ones at the site of injury that is necessary 337 to start tissue repair and bone regeneration. IL-4 has been reported to 338 stimulate proliferation and inhibit differentiation of osteoblastic cell 339 line [175]. Moreover, IL-4 overexpressing mice showed decreased 340 total bone mass and bone formation associated with decreased numbers 341 of differentiated osteoblasts [113]. IL-4 and IL-13 have also been report342 ed to inhibit prostaglandin synthesis in bone and stimulate migration of 343 osteoblasts [115,145]. Also, it has been shown that TGF-β and IL-10, 344 secreted by M2 macrophages, inhibit osteoclast formation and subse345 quent bone resorption [90,150]. 346 Generally, cytokines are not the only mediators of the inflammatory 347 response in bone tissue, other factors are also released in response to 348 inflammation. One important factor is osteoactivin (OA) that have 349 been discovered in our laboratory and was reported to stimulate MSC 350 differentiation into osteoblasts with increased mineralization of 351 the extracellular matrix (ECM), leading to significant bone growth 352 [1–3,5,147,161]. In addition, our lab also shows that OA works as a neg353 ative regulator of osteoclastogenesis. 354 Other groups have designated different names to OA in different 355 species, such as glycoprotein nmb (Gpnmb) in melanoma cell lines 356 [104,141,152,172,177], and melanocytes [16,17], dendritic cell heparan 357 sulfate proteoglycan integrin dependent ligand (DC-HIL) in dendritic 358 and T-cells [57,163] and human hematopoietic growth factor inducible 359 neurokinin (HGFIN) in tumor cells [18]. 360 Previous studies reported on the several functions of osteoactivin 361 (OA) in regulating cell proliferation, adhesion, differentiation and 362 synthesis of ECM proteins in normal and pathological conditions 363 Q26 [1,6,10,16,17,57,85,104,111,122,134,140,142,147,152,159,161,163,172]. 364 Interestingly, OA is produced by inflammatory macrophages recruited to 365 the site of injury in several diseases. Previous studies showed that OA is 366 not only produced by macrophages surrounding non-viable adipocyes 367 in obese individuals [76], but also by tumor-associated macrophages in 368 a combination with other bio-active extracelllular matrix proteins 369 (ECM) [114]. Moreover, OA expression was increased in heart infilatrated 370 macrophages in a mouse model of dilated cardiomyopathy [153]. In a 371 separate study, OA upregulation in uremic macrophages was reported 372 in patients with chronic nephritis; an end-stage renal disease [148]. 373 Furthermore, patients with acute liver injury showed a high levels of OA 374 in inflammatory liver macrophages, suggesting a significant role of OA 375 Q27 in regulating the immune response [85].

D

326 327

E

6

All authors state that they have no conflict of interest.

Uncited reference [129]

Please cite this article as: S.M. Abdelmagid, et al., Role of inflammation in the aging bone..., Life Sci (2014), http://dx.doi.org/10.1016/ j.lfs.2014.11.011

414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 Q30 436 437 438 439 440 441 442 443

445

446 Q32 447

S.M. Abdelmagid et al. / Life Sciences xxx (2014) xxx–xxx

R

R

E

C

D

P

R O

O

F

[30] A.E. Barr, M.F. Barbe, B.D. Clark, Systemic inflammatory mediators contribute to widespread effects in work-related musculoskeletal disorders, Exerc. Sport Sci. Rev. 32 (2004) 135–142. [31] A.E. Barr, M.F. Barbe, B.D. Clark, Systemic inflammatory mediators contribute to widespread effects in work-related musculoskeletal disorders, Exerc. Sport Sci. Rev. 32 (2004) 135–142. [32] A.E. Barr, M.F. Barbe, B.D. Clark, Work-related musculoskeletal disorders of the hand and wrist: epidemiology, pathophysiology, and sensorimotor changes, J. Orthop. Sports Phys. Ther. 34 (2004) 610–627. [33] A.E. Barr, F.F. Safadi, I. Gorzelany, M. Amin, S.N. Popoff, M.F. Barbe, Repetitive, negligible force reaching in rats induces pathological overloading of upper extremity bones, J. Bone Miner. Res. Off. J. Am. Soc. Bone Miner. Res. 18 (2003) 2023–2032. [34] P.M. Bartold, R.I. Marshall, D.R. Haynes, Periodontitis and rheumatoid arthritis: a review, J. Periodontol. 76 (2005) 2066–2074. [35] J.A. Baugh, R. Bucala, Macrophage migration inhibitory factor, Crit. Care Med. 30 (2002) S27–S35. [36] B.P. Bernard, Musculoskeletal disorders and workplace factors: a critical review of epidemiologic evidence for work-related musculoskeletal disorders of the neck, upper extremity and low backhttp://www.cdc.gov/niosh/docs/97-141/ US Government Printing Office, Washington, DC, USA, 1997. DHHS (NIOSH) Publication Number 97–141. [37] S.K. Biswas, M. Chittezhath, I.N. Shalova, J.Y. Lim, Macrophage polarization and plasticity in health and disease, Immunol. Res. 53 (2012) 11–24. [38] S.K. Biswas, A. Mantovani, Orchestration of metabolism by macrophages, Cell Metab. 15 (2012) 432–437. [39] L. Bjorklund, The Bone and Joint Decade 2000–2010. Inaugural meeting 17 and 18 April 1998, Lund, Sweden, Acta Orthop. Scand. Suppl. 281 (1998) 67–80. [40] A.L. Brubaker, J.L. Palmer, E.J. Kovacs, Age-related dysregulation of inflammation and innate immunity: lessons learned from rodent models, Aging Dis. 2 (2011) 346–360. [41] H. Bruunsgaard, B.K. Pedersen, Age-related inflammatory cytokines and disease, Immunol. Allergy Clin. N. Am. 23 (2003) 15–39. [42] T.W. Buford, M.D. Roberts, T.S. Church, Toward exercise as personalized medicine, Sports Med. 43 (2013) 157–165. [43] Bureau of Labor Statistics, Nonfatal occupational injuries and illnesses requiring days away from work, 2011http://www.bls.gov/news.release/osh2.nr0.htm News Release USDL-12–2204, November 8, 2012 2012. [44] N. Byl, F. Wilson, M. Merzenich, M. Melnick, P. Scott, A. Oakes, et al., Sensory dysfunction associated with repetitive strain injuries of tendinitis and focal hand dystonia: a comparative study, J. Orthop. Sports Phys. Ther. 23 (1996) 234–244. [45] E. Canalis, Interleukin-1 has independent effects on deoxyribonucleic acid and collagen synthesis in cultures of rat calvariae, Endocrinology 118 (1986) 74–81. [46] M.P. Cancro, Y. Hao, J.L. Scholz, R.L. Riley, D. Frasca, D.K. Dunn-Walters, et al., B cells and aging: molecules and mechanisms, Trends Immunol. 30 (2009) 313–318. [47] J.G. Cannon, M.A. Fiatarone, R.A. Fielding, W.J. Evans, Aging and stress-induced changes in complement activation and neutrophil mobilization, J. Appl. Physiol. 76 (1994) 2616–2620. [48] J.G. Cannon, R.A. Fielding, M.A. Fiatarone, S.F. Orencole, C.A. Dinarello, W.J. Evans, Increased interleukin 1 beta in human skeletal muscle after exercise, Am. J. Phys. 257 (1989) R451–R455. [49] M. Cantini, M.L. Massimino, A. Bruson, C. Catani, L. Dalla Libera, U. Carraro, Macrophages regulate proliferation and differentiation of satellite cells, Biochem. Biophys. Res. Commun. 202 (1994) 1688–1696. [50] S.J. Carp, M.F. Barbe, K.A. Winter, M. Amin, A.E. Barr, Inflammatory biomarkers increase with severity of upper-extremity overuse disorders, Clin. Sci. (Lond.) 112 (2007) 305–314. [51] S.J. Carp, M.F. Barbe, K.A. Winter, M. Amin, A.E. Barr, Inflammatory biomarkers increase with severity of upper-extremity overuse disorders, Clin. Sci. (Lond.) 112 (2007) 305–314. [52] S.J. Carp, A.E. Barr, M.F. Barbe, Serum biomarkers as signals for risk and severity of repetitive stress injuries, Biomark. Med 2 (2008) 67–79. [53] M. Centrella, T.L. McCarthy, E. Canalis, Tumor necrosis factor-alpha inhibits collagen synthesis and alkaline phosphatase activity independently of its effect on deoxyribonucleic acid synthesis in osteoblast-enriched bone cell cultures, Endocrinology 123 (1988) 1442–1448. [54] S.S. Chang, C.O. Weiss, Q.L. Xue, L.P. Fried, Association between inflammatoryrelated disease burden and frailty: results from the Women's Health and Aging Studies (WHAS) I and II, Arch. Gerontol. Geriatr. 54 (2012) 9–15. [55] K. Chotinantakul, W. Leeanansaksiri, Hematopoietic stem cell development, niches, and signaling pathways, Bone Marrow Res. 2012 (2012) 270425. [56] H.Y. Chung, M. Cesari, S. Anton, E. Marzetti, S. Giovannini, A.Y. Seo, et al., Molecular inflammation: underpinnings of aging and age-related diseases, Ageing Res. Rev. 8 (2009) 18–30. [57] J.S. Chung, K. Sato, I.I. Dougherty, P.D. Cruz Jr., K. Ariizumi, DC-HIL is a negative regulator of T lymphocyte activation, Blood 109 (2007) 4320–4327. [58] B.D. Clark, T.A. Al-Shatti, A.E. Barr, M. Amin, M.F. Barbe, Performance of a highrepetition, high-force task induces carpal tunnel syndrome in rats, J. Orthop. Sports Phys. Ther. 34 (2004) 244–253. [59] B.D. Clark, A.E. Barr, F.F. Safadi, L. Beitman, T. Al-Shatti, M. Amin, et al., Median nerve trauma in a rat model of work-related musculoskeletal disorder, J. Neurotrauma 20 (2003) 681–695. [60] B.D. Clark, A.E. Barr, F.F. Safadi, L. Beitman, T. Al-Shatti, M. Amin, et al., Median nerve trauma in a rat model of work-related musculoskeletal disorder, J. Neurotrauma 20 (2003) 681–695. [61] J.O. Coq, A.E. Barr, F. Strata, M. Russier, D.M. Kietrys, M.M. Merzenich, et al., Peripheral and central changes combine to induce motor behavioral deficits in a moderate repetition task, Exp. Neurol. 220 (2009) 234–245.

E

T

[1] S.M. Abdelmagid, M.F. Barbe, I. Arango-Hisijara, T.A. Owen, S.N. Popoff, F.F. Safadi, Osteoactivin acts as downstream mediator of BMP-2 effects on osteoblast function, J. Cell. Physiol. 210 (2007) 26–37. [2] S.M. Abdelmagid, M.F. Barbe, M. Hadjiargyrou, T.A. Owen, R. Razmpour, S. Rehman, et al., Temporal and spatial expression of osteoactivin during fracture repair, J. Cell. Biochem. 111 (2010) 295–309. [3] S.M. Abdelmagid, M.F. Barbe, M.C. Rico, S. Salihoglu, I. Arango-Hisijara, A.H. Selim, et al., Osteoactivin, an anabolic factor that regulates osteoblast differentiation and function, Exp. Cell Res. 314 (2008) 2334–2351. [4] S.M. Abdelmagid, A.E. Barr, M. Rico, M. Amin, J. Litvin, S.N. Popoff, et al., Performance of repetitive tasks induces decreased grip strength and increased fibrogenic proteins in skeletal muscle: role of force and inflammation, PLoS One 7 (2012) e38359. [5] S.M. Abdelmagid, J.Y. Belcher, F.M. Moussa, S.L. Lababidi, G.R. Sondag, K.M. Novak, et al., Mutation in osteoactivin decreases bone formation in vivo and osteoblast differentiation in vitro, Am. J. Pathol. 184 (2014) 697–713. [6] H. Abe, H. Uto, Y. Takami, Y. Takahama, S. Hasuike, M. Kodama, et al., Transgenic expression of osteoactivin in the liver attenuates hepatic fibrosis in rats, Biochem. Biophys. Res. Commun. 356 (2007) 610–615. [7] M. Abe, K. Hiura, J. Wilde, K. Moriyama, T. Hashimoto, S. Ozaki, et al., Role for macrophage inflammatory protein (MIP)-1alpha and MIP-1beta in the development of osteolytic lesions in multiple myeloma, Blood 100 (2002) 2195–2202. [8] I.E. Adamopoulos, L. Danks, I. Itonaga, R.M. Locklin, A. Sabokbar, D.J. Ferguson, et al., Stimulation of osteoclast formation by inflammatory synovial fluid, Virchows Arch. 449 (2006) 69–77. [9] I.E. Adamopoulos, A. Sabokbar, B.P. Wordsworth, A. Carr, D.J. Ferguson, N.A. Athanasou, Synovial fluid macrophages are capable of osteoclast formation and resorption, J. Pathol. 208 (2006) 35–43. [10] J.H. Ahn, Y. Lee, C. Jeon, S.J. Lee, B.H. Lee, K.D. Choi, et al., Identification of the genes differentially expressed in human dendritic cell subsets by cDNA subtraction and microarray analysis, Blood 100 (2002) 1742–1754. [11] T. Al-Shatti, A.E. Barr, F.F. Safadi, M. Amin, M.F. Barbe, Increase in inflammatory cytokines in median nerves in a rat model of repetitive motion injury, J. Neuroimmunol. 167 (2005) 13–22. [12] K.A. Alexander, M.K. Chang, E.R. Maylin, T. Kohler, R. Muller, A.C. Wu, et al., Osteal macrophages promote in vivo intramembranous bone healing in a mouse tibial injury model, J. Bone Miner. Res. Off. J. Am. Soc. Bone Miner. Res. 26 (2011) 1517–1532. [13] L.C. Almekinders, A.J. Baynes, L.W. Bracey, An in vitro investigation into the effects of repetitive motion and nonsteroidal antiinflammatory medication on human tendon fibroblasts, Am. J. Sports Med. 23 (1995) 119–123. [14] L.C. Almekinders, T.C. Logan, Results following treatment of traumatic dislocations of the knee joint, Clin. Orthop. Relat. Res. (1992) 203–207. [15] K.C. Anderson, J.D. Shaughnessy Jr., B. Barlogie, J.L. Harousseau, G.D. Roodman, Multiple myeloma, Hematology Am. Soc. Hematol. Educ. Program. (2002) 214–240. [16] M.G. Anderson, R.T. Libby, M. Mao, I.M. Cosma, L.A. Wilson, R.S. Smith, et al., Genetic context determines susceptibility to intraocular pressure elevation in a mouse pigmentary glaucoma, BMC Biol. 4 (2006) 20. [17] M.G. Anderson, R.S. Smith, N.L. Hawes, A. Zabaleta, B. Chang, J.L. Wiggs, et al., Mutations in genes encoding melanosomal proteins cause pigmentary glaucoma in DBA/2J mice, Nat. Genet. 30 (2002) 81–85. [18] P.S. Bandari, J. Qian, G. Yehia, D.D. Joshi, P.B. Maloof, J. Potian, et al., Hematopoietic growth factor inducible neurokinin-1 type: a transmembrane protein that is similar to neurokinin 1 interacts with substance P, Regul. Pept. 111 (2003) 169–178. [19] M.E. Barbe, W.E. Scott, P.J. Kutschke, A simplified approach to the treatment of Duane's syndrome, Br. J. Ophthalmol. 88 (2004) 131–138. [20] M.F. Barbe, A.E. Barr, Inflammation and the pathophysiology of work-related musculoskeletal disorders, Brain Behav. Immun. 20 (2006) 423–429. [21] M.F. Barbe, A.E. Barr, Inflammation and the pathophysiology of work-related musculoskeletal disorders, Brain Behav. Immun. 20 (2006) 423–429. [22] M.F. Barbe, A.E. Barr, I. Gorzelany, M. Amin, J.P. Gaughan, F.F. Safadi, Chronic repetitive reaching and grasping results in decreased motor performance and widespread tissue responses in a rat model of MSD, J. Orthop. Res. 21 (2003) 167–176. [23] M.F. Barbe, A.E. Barr, I. Gorzelany, M. Amin, J.P. Gaughan, F.F. Safadi, Chronic repetitive reaching and grasping results in decreased motor performance and widespread tissue responses in a rat model of MSD, J. Orthop. Res. 21 (2003) 167–176. [24] M.F. Barbe, M.B. Elliott, S.M. Abdelmagid, M. Amin, S.N. Popoff, F.F. Safadi, et al., Serum and tissue cytokines and chemokines increase with repetitive upper extremity tasks, J. Orthop. Res. (2008). [25] M.F. Barbe, M.B. Elliott, S.M. Abdelmagid, M. Amin, S.N. Popoff, F.F. Safadi, et al., Serum and tissue cytokines and chemokines increase with repetitive upper extremity tasks, J. Orthop. Res. 26 (2008) 1320–1326. [26] M.F. Barbe, F. Safadi, S. Popoff, A.E. Barr, Dose–response relationship between reach repetition and indicators of inflammation and movement dysfunction in a rat model of work-related musculoskeletal disorder, Temple Univ. J. Orthop. Surg. Sports Med. (2007) 67–71. [27] A.E. Barr, M.F. Barbe, Pathophysiological tissue changes associated with repetitive movement: a review of the evidence, Phys. Ther. 82 (2002) 173–187. [28] A.E. Barr, M.F. Barbe, Inflammation reduces physiological tissue tolerance in the development of work-related musculoskeletal disorders, J. Electromyogr. Kinesiol. 14 (2004) 77–85. [29] A.E. Barr, M.F. Barbe, Inflammation reduces physiological tissue tolerance in the development of work-related musculoskeletal disorders, J. Electromyogr. Kinesiol. 14 (2004) 77–85.

N C O

449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 Q33 506 507 508 509 Q34 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 Q35 528 529 530 531 532

References

U

448

7

Please cite this article as: S.M. Abdelmagid, et al., Role of inflammation in the aging bone..., Life Sci (2014), http://dx.doi.org/10.1016/ j.lfs.2014.11.011

533 Q36 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 Q37 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 Q2 613 614 615 616 617 618

D

P

R O

O

F

[93] HSE, The Health and Safety Executive Statistics 2010/11http://www.hse.gov.uk/ statistics/overall/hssh1011.pdf 2011. [94] R.L. Jilka, R.S. Weinstein, T. Bellido, A.M. Parfitt, S.C. Manolagas, Osteoblast programmed cell death (apoptosis): modulation by growth factors and cytokines, J. Bone Miner. Res. Off. J. Am. Soc. Bone Miner. Res. 13 (1998) 793–802. [95] A. Kawakami, K. Eguchi, N. Matsuoka, M. Tsuboi, T. Koji, S. Urayama, et al., Fas and Fas ligand interaction is necessary for human osteoblast apoptosis, J. Bone Miner. Res. Off. J. Am. Soc. Bone Miner. Res. 12 (1997) 1637–1646. [96] D.M. Kietrys, A.E. Barr-Gillespie, M. Amin, C.K. Wade, S.N. Popoff, M.F. Barbe, Aging contributes to inflammation in upper extremity tendons and declines in forelimb agility in a rat model of upper extremity overuse, PLoS One 7 (2012) e46954. [97] D.M. Kietrys, A.E. Barr, M.F. Barbe, Exposure to repetitive tasks induces motor changes related to skill acquisition and inflammation in rats, J. Mot. Behav. 43 (2011) 465–476. [98] H.R. Kim, K.W. Kim, H.G. Jung, K.S. Yoon, H.J. Oh, M.L. Cho, et al., Macrophage migration inhibitory factor enhances osteoclastogenesis through upregulation of RANKL expression from fibroblast-like synoviocytes in patients with rheumatoid arthritis, Arthritis Res. Ther. 13 (2011) R43. [99] J.H. Kim, H.M. Jin, K. Kim, I. Song, B.U. Youn, K. Matsuo, et al., The mechanism of osteoclast differentiation induced by IL-1, J. Immunol. 183 (2009) 1862–1870. [100] P. Kleinbongard, G. Heusch, R. Schulz, TNFalpha in atherosclerosis, myocardial ischemia/reperfusion and heart failure, Pharmacol. Ther. 127 (2010) 295–314. [101] Y.Y. Kong, U. Feige, I. Sarosi, B. Bolon, A. Tafuri, S. Morony, et al., Activated T cells regulate bone loss and joint destruction in adjuvant arthritis through osteoprotegerin ligand, Nature 402 (1999) 304–309. [102] R. Koopman, L.J. van Loon, Aging, exercise, and muscle protein metabolism, J. Appl. Physiol. 106 (2009) 2040–2048. [103] S. Kotake, T. Yago, M. Kawamoto, Y. Nanke, Role of osteoclasts and interleukin-17 in the pathogenesis of rheumatoid arthritis: crucial ‘human osteoclastology’, J. Bone Miner. Metab. 30 (2012) 125–135. [104] C.T. Kuan, K. Wakiya, J.M. Dowell, J.E. Herndon II, D.A. Reardon, M.W. Graner, et al., Glycoprotein nonmetastatic melanoma protein B, a potential molecular therapeutic target in patients with glioblastoma multiforme, Clin. Cancer Res. 12 (2006) 1970–1982. [105] J.I. Kuiper, J.H. van Dieen, V. Everts, J.H. Verbeek, M.H. Frings-Dresen, Associations between serum markers of collagen metabolism and spinal shrinkage, Clin. Biomech. (Bristol, Avon). 19 (2004) 209–212. [106] J.I. Kuiper, J.H. Verbeek, V. Everts, J.P. Straub, M.H. Frings-Dresen, Serum markers of collagen metabolism: construction workers compared to sedentary workers, Occup. Environ. Med. 62 (2005) 363–367. [107] J.I. Kuiper, J.H. Verbeek, J.P. Straub, V. Everts, M.H. Frings-Dresen, Physical workload of student nurses and serum markers of collagen metabolism, Scand. J. Work Environ. Health 28 (2002) 168–175. [108] H. Kuno, S.M. Kurian, G.N. Hendy, J. White, H.F. deLuca, C.O. Evans, et al., Inhibition of 1,25-dihydroxyvitamin D3 stimulated osteocalcin gene transcription by tumor necrosis factor-alpha: structural determinants within the vitamin D response element, Endocrinology 134 (1994) 2524–2531. [109] S.J. Leibovich, P.J. Polverini, H.M. Shepard, D.M. Wiseman, V. Shively, N. Nuseir, Macrophage-induced angiogenesis is mediated by tumour necrosis factor-alpha, Nature 329 (1987) 630–632. [110] S.X. Leng, J.A. Elias, Interleukin-11 inhibits macrophage interleukin-12 production, J. Immunol. 159 (1997) 2161–2168. [111] V. Lennerz, M. Fatho, C. Gentilini, R.A. Frye, A. Lifke, D. Ferel, et al., The response of autologous T cells to a human melanoma is dominated by mutated neoantigens, Proc. Natl. Acad. Sci. U. S. A. 102 (2005) 16013–16018. [112] S. Lentzsch, M. Gries, M. Janz, R. Bargou, B. Dorken, M.Y. Mapara, Macrophage inflammatory protein 1-alpha (MIP-1 alpha) triggers migration and signaling cascades mediating survival and proliferation in multiple myeloma (MM) cells, Blood 101 (2003) 3568–3573. [113] D.B. Lewis, H.D. Liggitt, E.L. Effmann, S.T. Motley, S.L. Teitelbaum, K.J. Jepsen, et al., Osteoporosis induced in mice by overproduction of interleukin 4, Proc. Natl. Acad. Sci. U. S. A. 90 (1993) 11618–11622. [114] M. Liguori, G. Solinas, G. Germano, A. Mantovani, P. Allavena, Tumor-associated macrophages as incessant builders and destroyers of the cancer stroma, Cancers 3 (2011) 3740–3761. [115] M. Lind, B. Deleuran, H. Yssel, E. Fink-Eriksen, K. Thestrup-Pedersen, IL-4 and IL-13, but not IL-10, are chemotactic factors for human osteoblasts, Cytokine 7 (1995) 78–82. [116] J. Lloberas, A. Celada, Effect of aging on macrophage function, Exp. Gerontol. 37 (2002) 1325–1331. [117] S. Mahbub, C.R. Deburghgraeve, E.J. Kovacs, Advanced age impairs macrophage polarization, J. Interf. Cytokine Res. 32 (2012) 18–26. [118] K. Mareschi, I. Ferrero, D. Rustichelli, S. Aschero, L. Gammaitoni, M. Aglietta, et al., Expansion of mesenchymal stem cells isolated from pediatric and adult donor bone marrow, J. Cell. Biochem. 97 (2006) 744–754. [119] A.C. Maue, E.J. Yager, S.L. Swain, D.L. Woodland, M.A. Blackman, L. Haynes, T-cell immunosenescence: lessons learned from mouse models of aging, Trends Immunol. 30 (2009) 301–305. [120] S. Maxson, E.A. Lopez, D. Yoo, A. Danilkovitch-Miagkova, M.A. Leroux, Concise review: role of mesenchymal stem cells in wound repair, Stem Cells Transl. Med. 1 (2012) 142–149. [121] P.L. McNeil, R. Khakee, Disruptions of muscle fiber plasma membranes. Role in exercise-induced damage, Am. J. Pathol. 140 (1992) 1097–1109. [122] J.S. Mo, M.G. Anderson, M. Gregory, R.S. Smith, O.V. Savinova, D.V. Serreze, et al., By altering ocular immune privilege, bone marrow-derived cells pathogenically contribute to DBA/2J pigmentary glaucoma, J. Exp. Med. 197 (2003) 1335–1344.

N

C

O

R

R

E

C

T

[62] C.W. Cotman, N.C. Berchtold, L.A. Christie, Exercise builds brain health: key roles of growth factor cascades and inflammation, Trends Neurosci. 30 (2007) 464–472. [63] T.N. Crotti, M.D. Smith, H. Weedon, M.J. Ahern, D.M. Findlay, M. Kraan, et al., Receptor activator NF-kappaB ligand (RANKL) expression in synovial tissue from patients with rheumatoid arthritis, spondyloarthropathy, osteoarthritis, and from normal patients: semiquantitative and quantitative analysis, Ann. Rheum. Dis. 61 (2002) 1047–1054. [64] J.C. Davis, M.C. Robertson, M.C. Ashe, T. Liu-Ambrose, K.M. Khan, C.A. Marra, International comparison of cost of falls in older adults living in the community: a systematic review, Osteoporos. Int. 21 (2010) 1295–1306. [65] R.J. Diaz-Garcia, T. Oda, M.J. Shauver, K.C. Chung, A systematic review of outcomes and complications of treating unstable distal radius fractures in the elderly, J. Hand Surg. 36 (2011) 824–835 e2. [66] J.B. Driban, A.E. Barr, M. Amin, M.R. Sitler, M.F. Barbe, Joint inflammation and early degeneration induced by high-force reaching are attenuated by ibuprofen in an animal model of work-related musculoskeletal disorder, J. Biomed. Biotechnol. 2011 (2011) 691412. [67] J.B. Driban, A.E. Barr, M. Amin, M.R. Sitler, M.F. Barbe, Joint inflammation and early degeneration induced by high-force reaching are attenuated by ibuprofen in an animal model of work-related musculoskeletal disorder, J. Biomed. Biotechnol. 2011 (2011) 691412. [68] M.B. Elliott, A.E. Barr, M.F. Barbe, Pro-nociceptive neurochemicals increase in spinal cord with high repetition reaching, Neurosci. Lett. 454 (2009) 33–37. [69] M.B. Elliott, A.E. Barr, B.D. Clark, M. Amin, S. Amin, M.F. Barbe, High force reaching task induces widespread inflammation, increased spinal cord neurochemicals and neuropathic pain, Neuroscience 158 (2009) 922–931. [70] M.B. Elliott, A.E. Barr, B.D. Clark, C.K. Wade, M.F. Barbe, Performance of a repetitive task by aged rats leads to median neuropathy and spinal cord inflammation with associated sensorimotor declines, Neuroscience 170 (2010) 929–941. [71] M.B. Elliott, A.E. Barr, D.M. Kietrys, T. Al-Shatti, M. Amin, M.F. Barbe, Peripheral neuritis and increased spinal cord neurochemicals are induced in a model of repetitive motion injury with low force and repetition exposure, Brain Res. 1218 (2008) 103–113. [72] M.B. Elliott, J.J. Jallo, R.F. Tuma, An investigation of cerebral edema and injury volume assessments for controlled cortical impact injury, J. Neurosci. Methods 168 (2008) 320–324. [73] G. Ferraccioli, A. Carbonella, E. Gremese, S. Alivernini, Rheumatoid arthritis and Alzheimer's disease: genetic and epigenetic links in inflammatory regulation, Discov. Med. 14 (2012) 379–388. [74] D.O. Freytes, J.W. Kang, I. Marcos-Campos, G. Vunjak-Novakovic, Macrophages modulate the viability and growth of human mesenchymal stem cells, J. Cell. Biochem. 114 (2013) 220–229. [75] C. Gabay, I. Kushner, Acute-phase proteins and other systemic responses to inflammation, N. Engl. J. Med. 340 (1999) 448–454. [76] T.L. Gabriel, M.J. Tol, R. Ottenhof, C. van Roomen, J. Aten, N. Claessen, et al., Lysosomal stress in obese adipose tissue macrophages contributes to MITFdependent GPNMB induction, Diabetes (2014). [77] R.L. Gallo, R.A. Dorschner, S. Takashima, M. Klagsbrun, E. Eriksson, M. Bernfield, Endothelial cell surface alkaline phosphatase activity is induced by IL-6 released during wound repair, J. Investig. Dermatol. 109 (1997) 597–603. [78] P. Geusens, W.F. Lems, Osteoimmunology and osteoporosis, Arthritis Res. Ther. 13 (2011) 242. [79] L. Gilbert, X. He, P. Farmer, S. Boden, M. Kozlowski, J. Rubin, et al., Inhibition of osteoblast differentiation by tumor necrosis factor-alpha, Endocrinology 141 (2000) 3956–3964. [80] L. Gilbert, X. He, P. Farmer, J. Rubin, H. Drissi, A.J. van Wijnen, et al., Expression of the osteoblast differentiation factor RUNX2 (Cbfa1/AML3/Pebp2alpha A) is inhibited by tumor necrosis factor-alpha, J. Biol. Chem. 277 (2002) 2695–2701. [81] S.R. Goldring, Pathogenesis of bone erosions in rheumatoid arthritis, Curr. Opin. Rheumatol. 14 (2002) 406–410. [82] S. Gordon, Alternative activation of macrophages, Nat. Rev. Immunol. 3 (2003) 23–35. [83] S. Gordon, The macrophage: past, present and future, Eur. J. Immunol. 37 (Suppl. 1) (2007) S9–17. [84] T.R. Hales, P.K. Bertsche, Management of upper extremity cumulative trauma disorders, AAOHN J. 40 (1992) 118–128. [85] B. Haralanova-Ilieva, G. Ramadori, T. Armbrust, Expression of osteoactivin in rat and human liver and isolated rat liver cells, J. Hepatol. 42 (2005) 565–572. [86] M. Hashizume, M. Mihara, The roles of interleukin-6 in the pathogenesis of rheumatoid arthritis, Arthritis 2011 (2011) 765624. [87] H. Hata, Bone lesions and macrophage inflammatory protein-1 alpha (MIP-1a) in human multiple myeloma, Leuk. Lymphoma 46 (2005) 967–972. [88] A.K. Hogasen, T.G. Abrahamsen, Polymyxin B stimulates production of complement components and cytokines in human monocytes, Antimicrob. Agents Chemother. 39 (1995) 529–532. [89] M. Holecek, Muscle wasting in animal models of severe illness, Int. J. Exp. Pathol. 93 (2012) 157–171. [90] M.H. Hong, H. Williams, C.H. Jin, J.W. Pike, The inhibitory effect of interleukin-10 on mouse osteoclast formation involves novel tyrosine-phosphorylated proteins, J. Bone Miner. Res. Off. J. Am. Soc. Bone Miner. Res. 15 (2000) 911–918. [91] R. Horton, GBD 2010: understanding disease, injury, and risk, Lancet 380 (2012) 2053–2054. [92] H. Hotokezaka, E. Sakai, N. Ohara, Y. Hotokezaka, C. Gonzales, K. Matsuo, et al., Molecular analysis of RANKL-independent cell fusion of osteoclast-like cells induced by TNF-alpha, lipopolysaccharide, or peptidoglycan, J. Cell. Biochem. 101 (2007) 122–134.

U

619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 Q5 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704

S.M. Abdelmagid et al. / Life Sciences xxx (2014) xxx–xxx

E

8

Please cite this article as: S.M. Abdelmagid, et al., Role of inflammation in the aging bone..., Life Sci (2014), http://dx.doi.org/10.1016/ j.lfs.2014.11.011

705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790

S.M. Abdelmagid et al. / Life Sciences xxx (2014) xxx–xxx

N C O

R

R

E

C

D

P

R O

O

F

[150] J. Pfeilschifter, S.M. Seyedin, G.R. Mundy, Transforming growth factor beta inhibits bone resorption in fetal rat long bone cultures, J. Clin. Invest. 82 (1988) 680–685. [151] J. Plowden, M. Renshaw-Hoelscher, C. Engleman, J. Katz, S. Sambhara, Innate immunity in aging: impact on macrophage function, Aging Cell. 3 (2004) 161–167. [152] V.A. Pollack, E. Alvarez, K.F. Tse, M.Y. Torgov, S. Xie, S.G. Shenoy, et al., Treatment parameters modulating regression of human melanoma xenografts by an antibody-drug conjugate (CR011-vcMMAE) targeting GPNMB, Cancer Chemother. Pharmacol. 60 (2007) 423–435. [153] S. Psarras, M. Mavroidis, D. Sanoudou, C.H. Davos, G. Xanthou, A.E. Varela, et al., Regulation of adverse remodelling by osteopontin in a genetic heart failure model, Eur. Heart J. 33 (2012) 1954–1963. [154] L. Punnett, D.H. Wegman, Work-related musculoskeletal disorders: the epidemiologic evidence and the debate, J. Electromyogr. Kinesiol. 14 (2004) 13–23. [155] S. Rani, M.F. Barbe, A.E. Barr, J. Litivn, Role of TNF alpha and PLF in bone remodeling in a rat model of repetitive reaching and grasping, J. Cell. Physiol. 225 (2010) 152–167. [156] S. Rani, M.F. Barbe, A.E. Barr, J. Litvin, The pattern of periostin-like factor and periostin in an animal model of work-related musculoskeletal disorder, Bone 44 (2009) 502–512. [157] S. Rani, M.F. Barbe, A.E. Barr, J. Litvin, Periostin-like-factor and periostin in an animal model of work-related musculoskeletal disorder, Bone 44 (2009) 502–512. [158] M. Rechardt, R. Shiri, S. Matikainen, E. Viikari-Juntura, J. Karppinen, H. Alenius, Soluble IL-1RII and IL-18 are associated with incipient upper extremity soft tissue disorders, Cytokine 54 (2011) 149–153. [159] J.N. Rich, Q. Shi, M. Hjelmeland, T.J. Cummings, C.T. Kuan, D.D. Bigner, et al., Bonerelated genes expressed in advanced malignancies induce invasion and metastasis in a genetically defined human cancer model, J. Biol. Chem. 278 (2003) 15951–15957. [160] S. Riondino, F. La Farina, F. Martini, F. Guadagni, P. Ferroni, Functional impairment in video terminal operators is related to low-grade inflammation, Int. Arch. Occup. Environ. Health 84 (2011) 745–751. [161] F.F. Safadi, J. Xu, S.L. Smock, M.C. Rico, T.A. Owen, S.N. Popoff, Cloning and characterization of osteoactivin, a novel cDNA expressed in osteoblasts, J. Cell. Biochem. 84 (2001) 12–26. [162] G.M. Shearer, Th1/Th2 changes in aging, Mech. Ageing Dev. 94 (1997) 1–5. [163] S. Shikano, M. Bonkobara, P.K. Zukas, K. Ariizumi, Molecular cloning of a dendritic cell-associated transmembrane protein, DC-HIL, that promotes RGD-dependent adhesion of endothelial cells through recognition of heparan sulfate proteoglycans, J. Biol. Chem. 276 (2001) 8125–8134. [164] T. Singh, A.B. Newman, Inflammatory markers in population studies of aging, Ageing Res. Rev. 10 (2011) 319–329. [165] P. Stashenko, F.E. Dewhirst, M.L. Rooney, L.A. Desjardins, J.D. Heeley, Interleukin-1 beta is a potent inhibitor of bone formation in vitro, J. Bone Miner. Res. Off. J. Am. Soc. Bone Miner. Res. 2 (1987) 559–565. [166] J.A. Stevens, P.S. Corso, E.A. Finkelstein, T.R. Miller, The costs of fatal and non-fatal falls among older adults, Inj. Prev. 12 (2006) 290–295. [167] R.D. Stout, C. Jiang, B. Matta, I. Tietzel, S.K. Watkins, J. Suttles, Macrophages sequentially change their functional phenotype in response to changes in microenvironmental influences, J. Immunol. 175 (2005) 342–349. [168] R.D. Stout, J. Suttles, Immunosenescence and macrophage functional plasticity: dysregulation of macrophage function by age-associated microenvironmental changes, Immunol. Rev. 205 (2005) 60–71. [169] T.T. Suh, K.W. Lyles, Osteoporosis considerations in the frail elderly, Curr. Opin. Rheumatol. 15 (2003) 481–486. [170] T. Tamura, N. Udagawa, N. Takahashi, C. Miyaura, S. Tanaka, Y. Yamada, et al., Soluble interleukin-6 receptor triggers osteoclast formation by interleukin 6, Proc. Natl. Acad. Sci. U. S. A. 90 (1993) 11924–11928. [171] R.P. Tracy, Emerging relationships of inflammation, cardiovascular disease and chronic diseases of aging, Int. J. Obes. Relat. Metab. Disord. 27 (Suppl. 3) (2003) S29–S34. [172] K.F. Tse, M. Jeffers, V.A. Pollack, D.A. McCabe, M.L. Shadish, N.V. Khramtsov, et al., CR011, a fully human monoclonal antibody-auristatin E conjugate, for the treatment of melanoma, Clin. Cancer Res. 12 (2006) 1373–1382. [173] P. Uciechowski, L. Kahmann, B. Plumakers, M. Malavolta, E. Mocchegiani, G. Dedoussis, et al., TH1 and TH2 cell polarization increases with aging and is modulated by zinc supplementation, Exp. Gerontol. 43 (2008) 493–498. [174] N. Udagawa, N. Takahashi, E. Jimi, K. Matsuzaki, T. Tsurukai, K. Itoh, et al., Osteoblasts/stromal cells stimulate osteoclast activation through expression of osteoclast differentiation factor/RANKL but not macrophage colonystimulating factor: receptor activator of NF-kappa B ligand, Bone 25 (1999) 517–523. [175] K. Ura, I. Morimoto, K. Watanabe, K. Saito, N. Yanagihara, S. Eto, Interleukin (IL)-4 and IL-13 inhibit the differentiation of murine osteoblastic MC3T3-E1 cells, Endocr. J. 47 (2000) 293–302. [176] C.Q. Wang, K.B. Udupa, H. Xiao, D.A. Lipschitz, Effect of age on marrow macrophage number and function, Aging 7 (1995) 379–384. [177] M.A. Weterman, N. Ajubi, I.M. van Dinter, W.G. Degen, G.N. van Muijen, D.J. Ruitter, et al., nmb, a novel gene, is expressed in low-metastatic human melanoma cell lines and xenografts, Int. J. Cancer J. Int. Cancer 60 (1995) 73–81. [178] B.R. Wong, D. Besser, N. Kim, J.R. Arron, M. Vologodskaia, H. Hanafusa, et al., TRANCE, a TNF family member, activates Akt/PKB through a signaling complex involving TRAF6 and c-Src, Mol. Cell 4 (1999) 1041–1049.

E

T

[123] D.E. Moller, Potential role of TNF-alpha in the pathogenesis of insulin resistance and type 2 diabetes, Trends Endocrinol. Metab. 11 (2000) 212–217. [124] T. Montalcini, S. Romeo, Y. Ferro, V. Migliaccio, C. Gazzaruso, A. Pujia, Osteoporosis in chronic inflammatory disease: the role of malnutrition, Endocrine 43 (2013) 59–64. [125] B.S. Moonga, O.A. Adebanjo, H.J. Wang, S. Li, X.B. Wu, B. Troen, et al., Differential effects of interleukin-6 receptor activation on intracellular signaling and bone resorption by isolated rat osteoclasts, J. Endocrinol. 173 (2002) 395–405. [126] T.I. Morales, V.C. Hascall, Effects of interleukin-1 and lipopolysaccharides on protein and carbohydrate metabolism in bovine articular cartilage organ cultures, Connect. Tissue Res. 19 (1989) 255–275. [127] S. Mori, Contribution of bone quality to fracture risk, Clin. Calcium 14 (2004) 33–38. [128] G.R. Mundy, L.G. Raisz, R.A. Cooper, G.P. Schechter, S.E. Salmon, Evidence for the secretion of an osteoclast stimulating factor in myeloma, N. Engl. J. Med. 291 (1974) 1041–1046. [129] P.J. Murray, T.A. Wynn, Obstacles and opportunities for understanding macrophage polarization, J. Leukoc. Biol. 89 (2011) 557–563. [130] P.J. Murray, T.A. Wynn, Protective and pathogenic functions of macrophage subsets, Nat. Rev. Immunol. 11 (2011) 723–737. [131] K.M. Musallam, F.S. Dahdaleh, A.I. Shamseddine, A.T. Taher, Incidence and prophylaxis of venous thromboembolic events in multiple myeloma patients receiving immunomodulatory therapy, Thromb. Res. 123 (2009) 679–686. [132] M.G. Nair, D.W. Cochrane, J.E. Allen, Macrophages in chronic type 2 inflammation have a novel phenotype characterized by the abundant expression of Ym1 and Fizz1 that can be partly replicated in vitro, Immunol. Lett. 85 (2003) 173–180. [133] M.G. Nair, I.J. Gallagher, M.D. Taylor, P. Loke, P.S. Coulson, R.A. Wilson, et al., Chitinase and Fizz family members are a generalized feature of nematode infection with selective upregulation of Ym1 and Fizz1 by antigen-presenting cells, Infect. Immun. 73 (2005) 385–394. [134] A. Nakamura, A. Ishii, C. Ohata, T. Komurasaki, Early induction of osteoactivin expression in rat renal tubular epithelial cells after unilateral ureteral obstruction, Exp. Toxicol. Pathol. 59 (2007) 53–59. [135] M.S. Nanes, J. Rubin, L. Titus, G.N. Hendy, B. Catherwood, Tumor necrosis factoralpha inhibits 1,25-dihydroxyvitamin D3-stimulated bone Gla protein synthesis in rat osteosarcoma cells (ROS 17/2.8) by a pretranslational mechanism, Endocrinology 128 (1991) 2577–2582. [136] C.F. Nathan, Macrophage activation: some questions, Ann. Ins. Pasteur Immunol. 137C (1986) 345–351. [137] R.M. Nesse, G.C. Williams, Evolution and the origins of disease, Sci. Am. 279 (1998) 86–93. [138] NIOSH, in: D.F. Utterback, T.M. Schnorr (Eds.), Use of workers' compensation data for occupational safety and health: proceedings from June 2012 workshop, US Department of Health and Human Services, Public Health Service, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, DHHS (NIOSH), Cincinnati, OH, 2013, pp. 1–232 Publication No 2013– 147, 2013 May. [139] NRC&IOM NRCIoM, Musculoskeletal disorders and the workplace: low back and upper extremities, The National Academies Press, 2001. [140] T. Ogawa, T. Nikawa, H. Furochi, M. Kosyoji, K. Hirasaka, N. Suzue, et al., Osteoactivin upregulates expression of MMP-3 and MMP-9 in fibroblasts infiltrated into denervated skeletal muscle in mice, Am. J. Physiol. Cell Physiol. 289 (2005) C697–C707. [141] I. Okamoto, C. Pirker, M. Bilban, W. Berger, D. Losert, C. Marosi, et al., Seven novel and stable translocations associated with oncogenic gene expression in malignant melanoma, Neoplasia 7 (2005) 303–311. [142] M. Onaga, A. Ido, S. Hasuike, H. Uto, A. Moriuchi, K. Nagata, et al., Osteoactivin expressed during cirrhosis development in rats fed a choline-deficient, L-amino acid-defined diet, accelerates motility of hepatoma cells, J. Hepatol. 39 (2003) 779–785. [143] S. Onodera, J. Nishihira, K. Iwabuchi, Y. Koyama, K. Yoshida, S. Tanaka, et al., Macrophage migration inhibitory factor up-regulates matrix metalloproteinase-9 and -13 in rat osteoblasts. Relevance to intracellular signaling pathways, J. Biol. Chem. 277 (2002) 7865–7874. [144] S. Onodera, S. Sasaki, S. Ohshima, N. Amizuka, M. Li, N. Udagawa, et al., Transgenic mice overexpressing macrophage migration inhibitory factor (MIF) exhibit highturnover osteoporosis, J. Bone Miner. Res. Off. J. Am. Soc. Bone Miner. Res. 21 (2006) 876–885. [145] Y. Onoe, C. Miyaura, T. Kaminakayashiki, Y. Nagai, K. Noguchi, Q.R. Chen, et al., IL-13 and IL-4 inhibit bone resorption by suppressing cyclooxygenase-2dependent prostaglandin synthesis in osteoblasts, J. Immunol. 156 (1996) 758–764. [146] S. Oshima, S. Onodera, N. Amizuka, M. Li, K. Irie, S. Watanabe, et al., Macrophage migration inhibitory factor-deficient mice are resistant to ovariectomy-induced bone loss, FEBS Lett. 580 (2006) 1251–1256. [147] T.A. Owen, S.L. Smock, S. Prakash, L. Pinder, D. Brees, D. Krull, et al., Identification and characterization of the genes encoding human and mouse osteoactivin, Crit. Rev. Eukaryot. Gene Expr. 13 (2003) 205–220. [148] M.V. Pahl, N.D. Vaziri, J. Yuan, S.G. Adler, Upregulation of monocyte/macrophage HGFIN (Gpnmb/osteoactivin) expression in end-stage renal disease, Clin. J. Am. Soc. Nephrol. 5 (2010) 56–61. [149] J. Peake, P. Della Gatta, D. Cameron-Smith, Aging and its effects on inflammation in skeletal muscle at rest and following exercise-induced muscle injury, Am. J. Physiol. Regul. Integr. Comp. Physiol. 298 (2010) R1485–R1495.

U

791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876

9

Please cite this article as: S.M. Abdelmagid, et al., Role of inflammation in the aging bone..., Life Sci (2014), http://dx.doi.org/10.1016/ j.lfs.2014.11.011

877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 Q7 899 900 901 902 903 904 905 906 907 908 909 910 911 912 913 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959 960 961 962

10

[181] D.L. Xin, M.Y. Harris, C.K. Wade, M. Amin, A.E. Barr, M.F. Barbe, Aging enhances serum cytokine response but not task-induced grip strength declines in a rat model of work-related musculoskeletal disorders, BMC Musculoskelet. Disord. 12 (2011) 63. [182] H.S. Yeh, J.R. Berenson, Treatment for myeloma bone disease, Clin. Cancer Res. 12 (2006) 6279s–6284s.

N

C

O

R

R

E

C

T

E

D

P

R O

O

F

[179] A.D. Woolf, B. Pfleger, Burden of major musculoskeletal conditions, Bull. World Health Organ. 81 (2003) 646–656. [180] D.L. Xin, M.Y. Harris, C.K. Wade, M. Amin, A.E. Barr, M.F. Barbe, Aging enhances serum cytokine response but not task-induced grip strength declines in a rat model of work-related musculoskeletal disorders, BMC Musculoskelet. Disord. 12 (2011) 63.

U

963 964 965 966 967 968

S.M. Abdelmagid et al. / Life Sciences xxx (2014) xxx–xxx

Please cite this article as: S.M. Abdelmagid, et al., Role of inflammation in the aging bone..., Life Sci (2014), http://dx.doi.org/10.1016/ j.lfs.2014.11.011

969 970 971 Q9

Role of inflammation in the aging bones.

Chronic inflammation in aging is characterized by increased inflammatory cytokines, bone loss, decreased adaptation, and defective tissue repair in re...
1MB Sizes 0 Downloads 7 Views