You are on page 1of 9

Am J Physiol Regul Integr Comp Physiol 315: R104–R112, 2018.

First published March 28, 2018; doi:10.1152/ajpregu.00021.2018.

REVIEW 2017 APS Conference: Physiological and Pathophysiological


Consequences of Sickle Cell Disease

Targeting pain at its source in sickle cell disease


Kanika Gupta,* Om Jahagirdar,* and Kalpna Gupta
Vascular Biology Center, Division of Hematology, Oncology and Transplantation, Department of Medicine, University of
Minnesota, Minneapolis, Minnesota
Submitted 25 January 2018; accepted in final form 20 March 2018

Gupta K, Jahagirdar O, Gupta K. Targeting pain at its source in sickle cell


disease. Am J Physiol Regul Integr Comp Physiol 315: R104 –R112, 2018. First
published March 28, 2018; doi:10.1152/ajpregu.00021.2018.—Sickle cell disease
(SCD) is a genetic disorder associated with hemolytic anemia, end-organ damage,
reduced survival, and pain. One of the unique features of SCD is recurrent and
unpredictable episodes of acute pain due to vasoocclusive crisis requiring hospi-
talization. Additionally, patients with SCD often develop chronic persistent pain.
Currently, sickle cell pain is treated with opioids, an approach limited by adverse
effects. Because pain can start at infancy and continue throughout life, preventing
the genesis of pain may be relatively better than treating the pain once it has been
evoked. Therefore, we provide insights into the cellular and molecular mechanisms
of sickle cell pain that contribute to the activation of the somatosensory system in
the peripheral and central nervous systems. These mechanisms include mast cell
activation and neurogenic inflammation, peripheral nociceptor sensitization, mal-
adaptation of spinal signals, central sensitization, and modulation of neural circuits
in the brain. In this review, we describe potential preventive/therapeutic targets and
their targeting with novel pharmacologic and/or integrative approaches to amelio-
rate sickle cell pain.
analgesia; mast cell; neurogenic inflammation; opioid; pain; sickle cell disease;
substance P; vasoocclusive crises

INTRODUCTION patients with SCD may also experience chronic pain. Chronic
pain may be the result of maladaptive changes in pain path-
Sickle cell disease (SCD) affects millions of people world- ways. Current understanding of pain in SCD suggests that pain
wide (65). Although the molecular basis of this genetic disease occurs by nociceptive, inflammatory, and neuropathic mecha-
is a point mutation in the ␤-chain of hemoglobin, its conse- nisms (77). Emerging observations suggest several pain path-
quences are manyfold (7, 86). One of the major comorbidities ways that could be targeted to ameliorate or reduce pain.
of SCD is pain. In SCD, pain can start in infancy and continue In this review, we describe the mechanisms of acute and
throughout life, often leading to hospitalization and poor qual- chronic pain in SCD, focusing on how sickle pathobiology
ity of life (1, 77). Two types of pain have been characterized in evokes pain by activating the peripheral and central nervous
SCD: acute pain and chronic pain. Acute pain is associated systems (Fig. 1). We provide brief insight into the unique
with vasoocclusion. Under low oxygen tension, sickle hemo- humanized transgenic mouse models of SCD, which have been
globin polymerizes into rigid fibers and makes red blood cells critical in defining the mechanisms of sickle cell pain. We also
(RBCs) less deformable. Such stiff RBCs occlude microcircu- elaborate on the targeting of these mechanisms with specific
lation and cause vasoocclusive crisis (VOC) characterized by pharmacologic and/or integrative approaches with the potential
episodic, recurrent, and unpredictable acute pain (7, 8, 68). to treat sickle cell pain more effectively.
During VOC, there is impaired oxygenation of the distal tissue
HUMANIZED MOUSE MODELS OF SCD
and ischemic-reperfusion injury. This injury leads to inflam-
mation, oxidative stress, and endothelial dysfunction, resulting Transgenic mouse models of SCD expressing human sickle
in acute pain (1, 8). In addition to the acute pain of VOC, hemoglobin have played a key role in understanding the
pathobiology of pain in SCD, reviewed by Tran et al. (77).
There are two broad categories of such mice. The first type of
* Kanika Gupta and Om Jahagirdar contributed equally to this work. mice expresses human ␤-sickle hemoglobin alongside mouse
Address for reprint requests and other correspondence: Kalpna Gupta,
Vascular Biology Center, Medicine - Hematology, Oncology and Transplan-
␣- and ␤-globin; these include the NY1DD and S⫹SAntilles
tation, Univ. of Minnesota, Mayo Mail Code 480, 420 Delaware St., SE, mice with a mild-to-moderate SCD phenotype (29, 30). The
Minneapolis, MN 55455 (e-mail: gupta014@umn.edu). second type of mice expresses exclusively human ␣- and
R104 0363-6119/18 Copyright © 2018 the American Physiological Society http://www.ajpregu.org
Downloaded from journals.physiology.org/journal/ajpregu (103.092.202.027) on August 6, 2023.
TARGETING SICKLE CELL PAIN R105
effort between the Analgesic, Anesthetic, and Addiction Clin-
ical Trail Translations, Innovations, Opportunities, and Net-
works (ACTTION) and the American Pain Society has led to
an evidence-based classification system to facilitate the diag-
nosis and management of chronic sickle cell pain (24), the
ACTTION-American Pain Society Pain Taxonomy (AAPT).
These criteria may improve the diagnosis of chronic pain, but
new treatments are dependent on a better understanding of the
sickle pathobiology that perpetuates pain.
SICKLE PATHOBIOLOGY UNDERLYING THE ACTIVATION
OF SOMATOSENSORY MECHANISMS LEADING TO PAIN

The unique sickle pathobiology of vasoocclusion, inflamma-


tion, and oxidative stress along with ischemia-reperfusion
injury may create a noxious environment in the periphery,
spinal cord, and/or brain, contributing to a complex pathology
underlying pain in SCD (Fig. 1). Two key processes that may
drive chronic as well as acute pain are neurogenic inflamma-
Fig. 1. Pain pathways from the periphery to the brain and from the brain and
tion and mast cell activation.
central nervous system to the periphery. We propose that sickle pathobiology
replete with vasoocclusion, ischemia-reperfusion injury, inflammation, and Neurogenic Inflammation in SCD
oxidative stress activates mast cells, which in turn leads to peripheral nocice-
ptor sensitization. Signals from the periphery are transmitted to the dorsal horn Inflammation caused by nerve stimulation is called “neuro-
of the spinal cord via primary afferents, which are modulated and further genic inflammation” (36). It leads to the release of cytokines
transmitted to the brain. However, sickle cell disease pathobiology may and neuropeptides including substance P (SP) and calcitonin
influence each of these components of pain independently. Moreover, sus- gene-related peptide (CGRP) from nerve fibers, which stimu-
tained hyperexcitability of second-order neurons in the spinal cord may lead to
an antidromic release of action potentials and neurotransmitters from the
late vascular dilatation and increased venular permeability.
central nervous system to the periphery, leading to a feedback loop of Sickle BERK mice show neurogenic inflammation as evi-
peripheral and central sensitization. In addition to receiving signals from the denced by an increase in vascular leakage compared with
spinal cord, the brain may modulate pain by releasing neurotransmitters control BERK mice (84). This process may underlie enhanced
through the descending inhibitory pathway into the dorsal horn, which may peripheral nociceptor sensitization observed by us in these
have an inhibitory or facilitatory effect on pain. DRG, dorsal root ganglion.
mice (79) described in CENTRAL AND PERIPHERAL SENSITIZATION OF
CHRONIC PAIN PATHWAYS IN SCD. Significantly higher circulating
␤-globin chains and no mouse ␣- and ␤-globin chains. These SP has been described in patients with SCD at steady state
include the HbSS-BERK and HbSS-Townes sickle mice, compared with healthy controls (55). One of the sources of SP
which exclusively express ⬎99% human sickle hemoglobin appears to be mast cells as described below.
(HbS), and control HbAA-BERK or HbAA-Townes mice,
which express normal human hemoglobin (HbA; 64, 69). Mast Cell Activation in SCD
HbSS-BERK and HbSS-Townes mice constitutively show me- Mast cells are tissue-resident leukocytes, unlike neutrophils,
chanical, thermal, and deep tissue hyperalgesia, characteristic which are found in circulation (36). Mast cells display tremen-
features of chronic pain in patients with SCD (1, 17, 37, 38, 45, dous heterogeneity in their activation, which is specific to the
47, 56). Using hypoxia-reoxygenation treatments, we have surrounding microenvironment (36). An important feature of
been able to evoke acute pain in these sickle mice to simulate mast cell activation is their ability to release a variety of
pain due to VOC (17, 81). BERK sickle mice exhibit a substances by degranulation and/or de novo synthesis, includ-
significantly greater degree of hyperalgesia for all behavioral ing proteases, adhesion molecules, neuropeptides, and cyto-
measures compared with sex- and age-matched Townes sickle kines, which act in a paracrine as well as autocrine manner.
mice (39). These models have been highly instructive in Notably, mast cells also release extracellular traps, vesicles,
examining the pathobiology of sickle cell pain and show podia, and nanotubes, which physically interact directly with
translational potential. The genetically distinct “knock-in” other cellular structures. Since mast cells are located in the
strategy of human ␣- and ␤-transgene insertion in Townes vicinity of vasculature and nerve fibers, their activation has
mice compared with BERK mice may provide relative advan- direct influence on circulatory and neural physiology. We
tage for further genetic manipulations to examine specific found degranulating mast cells in the skin in close proximity to
mechanisms of pain (39). nerve fibers, particularly in the nerve plexus of BERK sickle
PATHOBIOLOGY OF CHRONIC PAIN IN SCD
mice (36, 84). We found that mast cells release tryptase, which
activates protease-activated receptor 2 (PAR2) and transient
Chronic pain is defined as pain that continues for ⬎3 mo receptor potential cation channel V1 (TRPV1) channels on
(14). A significant majority of patients with SCD develop nerve endings, leading to the release of neuropeptides SP and
chronic pain with increasing age (54, 73). Development of new CGRP. These neuropeptides act on the vasculature leading to
treatments is hampered by the long-term requirement of opi- increased permeability and neurogenic inflammation and a
oids, lack of objective pain measures, and relatively poor vicious cycle of mast cell activation and pain. Treatment of
understanding of symptoms and mechanisms. A collaborative sickle mice with imatinib, a c-kit inhibitor, or cromolyn so-

AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00021.2018 • www.ajpregu.org


Downloaded from journals.physiology.org/journal/ajpregu (103.092.202.027) on August 6, 2023.
R106 TARGETING SICKLE CELL PAIN

dium, a mast cell stabilizer, led to reduced inflammation, control BERK mice demonstrate a higher rate of spontaneous
neuroinflammation, and hyperalgesia. Deletion of mast cells discharge, enlarged receptive fields, reduced mechanical thresh-
from sickle mice ameliorated chronic pain; this finding sug- old, and prolonged after-discharges after mechanical stimulation
gests that mast cell activation contributes to pain. As described (20). These electrophysiological changes are accompanied by
below in MECHANISM-BASED STRATEGIES TO TARGET SICKLE CELL increased phosphorylation of the MAPK family members p42/44
PAIN, imatinib treatment completely ameliorated VOC in pa- MAPK, ERK, JNK, and p38 MAPK in the spinal cord, which is
tients with SCD (61, 75). It is likely that mast cell activation in consistent with chronic pain pathways (20).
SCD underlies the stimulation of selectins on the endothelial In addition to central sensitization, we observed sensitization
surface and activation of neutrophils, as described below, of cutaneous nociceptors in the periphery in sickle BERK mice
which orchestrate sickle RBC adhesion and vasoocclusion, as (79). Electrophysiological recordings of the tibial nerve in vivo
well as contribute to coagulopathy by activating platelets. in live anesthetized mice demonstrated spontaneous activity in
the A␦ and C fiber nociceptors. The C fibers showed increased
PATHOBIOLOGY OF ACUTE PAIN IN SCD response to mechanical and thermal stimuli in sickle BERK
compared with control BERK mice. Complementary to these
A characteristic and unique feature of SCD is unpredictable
observations, TRPV1 channels on the primary afferent termi-
and recurrent VOC due to occlusion of venules accompanied
by ischemia-reperfusion injury, inflammation, hemolysis, and nals and somata have been shown to be activated and contrib-
oxidative stress (8, 68, 73). Acute pain, considered worse than ute to mechanical hypersensitivity in sickle BERK mice (38).
the pain of childbirth, requires hospitalization and is often The molecular signatures of these electrophysiological out-
challenging to treat (8). Under low oxygen tension, sickle comes are emerging. We found that oxidative stress, SP, glial
erythrocyte HbS polymerizes into rigid fibers resulting in the activation, glial fibrillary acidic protein, and inflammation are
typical sickle shape of erythrocytes (16). Rigid sickle RBCs increased significantly in the dorsal horn of sickle BERK mice
aggregate and adhere to activated endothelium, along with compared with control BERK mice (84, 85). The spontaneous
circulating leukocytes and platelets (90, 92). Leukocytes, spe- nociceptor activity in the periphery and spinal cord and
cifically neutrophils, are activated in sickle mice and patients hypersensitivity to mechanical and/or thermal stimuli are
with SCD (50). These interactions of sickle RBC and leuko- suggestive of central sensitization in sickle mice, which may
cytes with vascular endothelium are facilitated by adhesion underlie chronic pain and be recalcitrant to analgesic ther-
molecules including E- and P-selectin (52, 66). Vasoocclusion apy. It is likely that this hyperexcitability of dorsal horn
is prevented in sickle mice lacking E- and P-selectin (78). neurons is a continuum of peripherally evoked sensory
Blockade of P-selectin with heparin improved microvascular activity and/or may be due to the glial activation by sickle
blood flow in patients with SCD (46, 52). Crizanlizumab, an pathobiology involving mast cells and ischemia-reperfusion
antibody to P-selectin, significantly reduced pain crises in injury in the spinal cord (Fig. 1).
patients with SCD in a phase II trial, described in detail in
MECHANISM-BASED STRATEGIES TO TARGET SICKLE CELL PAIN (6). NEUROMODULATORY PATHWAYS
However, it remains to be determined how vasoocclusion leads Signals in the dorsal horn of the spinal cord are also
to acute pain. modulated by neurotransmitters including dopamine, endoge-
Upregulation of selectins on the endothelial surface is crit- nous opioids, ␥-aminobutyric acid, glycine, and others released
ical for adhesion of sickle RBCs and leukocytes. Mast cell from the nerve fiber projections from the brain stem. These
activation induces P-selectin-dependent leukocyte rolling and top-down mechanisms originating in the brain may be influ-
adhesion in postcapillary venules in vivo (32, 76). Activation enced by perception and lead to affective modulation, which
of mast cells leads to endothelial leukocyte adhesion molecule may have an inhibitory or facilitatory effect on pain transmis-
(ELAM) expression on vasculature in the skin (44). In the sion. Neuroimaging studies in sickle cell subjects with chronic
Klein et al. (44) study, neonatal human foreskins, incubated pain have shown abnormal activity in two key neuronal net-
with morphine, a known activator of mast cell degranulation, works of the brain, namely, the default mode network (DMN)
led to increased ELAM expression. We observed extremely and executive control network (ECN; 19, 25). The DMN is a
large aggregates of activated mast cells releasing extracellular network of highly correlated areas of the brain that is active
traps in close association with the intact vasculature in the skin
when the brain is engaged in nonpurposeful actions, such as
of sickle mice (36). Thus, activation of mast cells in SCD may
daydreaming, not conscious and purposeful action. The key
play a critical role in VOC and acute pain in addition to chronic
zones of the DMN are the ventral medial prefrontal cortex, the
pain.
inferior parietal lobule, the posterior cingulate cortex, the
CENTRAL AND PERIPHERAL SENSITIZATION OF CHRONIC
precuneus, areas of the medial frontal gyri, the hippocampal
PAIN PATHWAYS IN SCD
formation, and the posterior lateral temporal cortex (15). In
patients with SCD, the neuronal circuits that enhance pain are
In chronic pain, constitutively hyperexcitable nociceptors on sensitized, and the circuits that suppress pain are desensitized.
peripheral afferents and/or second-order neurons in the spinal In SCD subjects with increased pain, pronociceptive connec-
cord continue to transmit action potentials on a sustained basis tions between the anterior cingulate cortex and areas of the
in the absence of noxious insult. Hypersensitivity to noxious DMN such as the precuneus and the inferior parietal lobule are
and innocuous stimuli— called “hyperalgesia” and “allodynia,” increased (25). In contrast, SCD subjects with infrequent pain
respectively— has been observed in sickle mice and patients show increased connectivity between the opioid-rich antinoci-
(12, 17, 18, 38, 42, 45, 47, 84). Electrophysiological recordings ceptive perigenual and subgenual cingulate and the DMN.
in the dorsal horn neurons of sickle BERK mice compared with There is also increased connectivity between the perigenual

AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00021.2018 • www.ajpregu.org


Downloaded from journals.physiology.org/journal/ajpregu (103.092.202.027) on August 6, 2023.
TARGETING SICKLE CELL PAIN R107
anterior cingulate area and the salience network (SLN; 25). viewed by Gupta et al. (37). Furthermore, opioids adversely
The SLN is the main area for pain processing and includes the influence RBC rheology by altering their membrane structure,
bilateral primary and secondary somatosensory cortexes, the increase mast cell degranulation-induced inflammation, and in-
anterior insula, the primary motor cortex, and the sensory fluence organ pathology via their coactivation of receptor tyrosine
motor area (9). The ECN is the part of the brain that is active kinases leading to mitogenic signaling (35, 37). Importantly,
when the brain is engaged in conscious, purposeful action. The opioids do not treat the underlying mechanisms evoking pain, and
ECN is involved in external processing such as nociception and considerable concerns regarding opioid use have led to opioid
decision making. The ECN comprises the dorsolateral prefron- phobia, often leading to undertreatment of sickle cell pain (8).
tal cortex and the posterior parietal cortex (53). In patients with Recent understanding of the mechanism-based targets of pain
SCD, this part of the brain is hyperactive and may contribute to described above has the potential to lead to improved analgesic
exaggerated pain perception by the patient. Sociopsychological therapies in SCD, described below (Table 1).
factors leading to cognitive impairment have been described to
influence pain and opioid requirement in patients with SCD Targeting VOC in SCD
(31). In addition, cerebral infarction in SCD subjects also During the initial phases of VOC, P-selectin initiates the
results in a significant decline of neuropsychological functions, adhesion of sickle erythrocytes and leukocytes to the endothe-
IQ, and cognitive score (5, 10, 13, 22, 26, 71, 87). However, lium and mediates the adhesion of activated platelets to neu-
molecular events underlying these neural processes in the brain trophils, resulting in vasoocclusion and inflammation (68). The
remain unknown. It is likely that local ischemia-reperfusion anti-P-selectin antibody, crizanlizumab, inhibits P-selectin-me-
injury, mast cell activation, and other inflammatory processes diated cell-cell adhesion and has been shown to reduce the
may directly influence the neural circuits and/or may be a result frequency of painful VOC by 45% and triple the median time of
of somatosensory inputs from the periphery. first VOC in a phase II clinical trial (6). Crizanlizumab may be the
first drug to be approved by the US Food and Drug Administra-
MECHANISM-BASED STRATEGIES TO TARGET SICKLE tion (FDA) to prevent VOC and subsequent pain at its source.
CELL PAIN
Inhibiting Inflammation
Opioids have been the mainstay of symptomatic manage-
ment of sickle cell pain. Use of opioids is fraught with side Elastase released from activated leukocytes mediates inflam-
effects including hyperalgesia, dependence, and tolerance, re- mation in the peripheral tissues and the dorsal root ganglion

Table 1. Mechanism-based therapies to target sickle cell pain


Intervention Mechanism Known Outcomes and Current Clinical Trials

Crizanlizumab Anti-P-selectin antibody Reduced the frequency of painful VOC by 45% and tripled the median time of first VOC in
patients with SCD; current clinical trial (ClinicalTrials.gov identifier NCT03264989:
“Pharmacokinetics and Pharmacodynamics Study of Crizanlizumab in Adult SCD Patients
with VOC”)
Sivelestat Leukocyte elastase inhibitor Decreased neuronal injury to the DRG and reduced neuropathic pain in BERK sickle mice
Imatinib cKIT/mast cell inhibitor Reduced neurogenic inflammation and prevented hypoxia-reoxygenation-induced hyperalgesia
in sickle mice; decreased frequency of VOC in patients with SCD
Cromolyn Mast cell stabilizer Increased the analgesic effect of a suboptimal dose of morphine in BERK sickle mice
Trifluoperazine CaMKII␣ inhibitor; reduces In a phase I clinical trial, ameliorated neurogenic pain and caused 50% reduction in chronic
calpain-1 activity pain in patients with SCD without severe sedation or supplemental opioid analgesics
Simvastatin Decreases calpain-1 activation? In patients with SCD, reduced frequency of pain, oral analgesic use, and markers of
inflammation, acting synergistically with hydroxyurea; 4 completed clinical trials are cited
on the ClinicalTrials.gov website
CP55,940 Cannabinoid receptor 1 and 2 Ameliorated chronic and hypoxia-reoxygenation-induced hyperalgesia in sickle mice; mitigated
agonist mast cell activation, inflammation, and neurogenic inflammation in sickle mice; current
clinical trial (ClinicalTrials.gov identifier NCT01771731: “Vaporized Cannabis for Chronic
Pain Associated with SCD”)
Rapamycin mTOR inhibitor: increases fetal Ameliorated the nociception phenotype in sickle mice; in 1 renal transplant recipient, increased
everolimus hemoglobin levels fetal hemoglobin levels from 4.8 to 15% and was well tolerated
Omega-3 fatty Antioxidant Reduced frequency of VOC and transfusion requirements in a randomized study of 140
acids patients in Sudan; current clinical trial (ClinicalTrials.gov identifier NCT02947100:
“Omega–3 Fatty Acids in Sickle Cell Disease”)
Curcumin and Anti-inflammatory and antioxidant Attenuated glial activation, neuroinflammation, and oxidative stress in spinal cords of BERK
CoQ10 sickle mice, resulting in attenuation of hyperalgesia; reduced inflammation, oxidative stress,
and VOC-associated pain in patients with SCD
AT-200 Nociceptin opioid receptor agonist Ameliorated chronic and hypoxia-reoxygenation-induced mechanical, thermal, and deep tissue/
and mast cell inhibitor musculoskeletal hyperalgesia in BERK sickle mice without causing tolerance
Acupuncture Inhibits inflammation peripherally In awake BERK sickle mice, reduced inflammatory cytokines, substance P, and neurogenic
and in the spinal cord inflammation in the periphery and signaling pathways of nociception in the spinal cord and
potentiated the effect of a suboptimal dose of morphine; acupuncture significantly reduced
VOC-associated pain in patients with SCD
Hypnosis Modulates vascular physiology Decreased pain intensity and increased peripheral blood flow during anticipation and
experience of pain in patients with SCD
CoQ10, coenzyme Q10; DRG, dorsal root ganglion; mTOR, mammalian target of rapamycin; SCD, sickle cell disease; VOC, vasoocclusive crisis.

AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00021.2018 • www.ajpregu.org


Downloaded from journals.physiology.org/journal/ajpregu (103.092.202.027) on August 6, 2023.
R108 TARGETING SICKLE CELL PAIN

(DRG), contributing to pain. Vicuña et al. showed that upon the sole measure of intensity of pain in this study, may not have
peripheral nerve injury in rodents, elastase is released from fully captured the multidimensional nature of intensity of pain
infiltrating T cells in the DRG, contributing to neuropathic pain in SCD (39).
[82a; see Weyer and Stucky (88)]. SerpinA3N, an endogenous
inhibitor of serine proteases, and the drug sivelestat, a leuko- Stimulating Cannabinoid Receptors
cyte elastase inhibitor, alleviated pain in this mouse model of
neuropathic pain as measured by the paw withdrawal threshold Cannabinoid receptor type 1 (CB1R) and cannabinoid re-
and responses to von Frey filaments (P ⬍ 0.05). We found that ceptor type 2 (CB2R) activation on mast cells has been shown
leukocyte elastase activity is significantly increased in the to inhibit degranulation and inflammation, respectively (72).
DRG and lungs of sickle mice (2). Sivelestat treatment de- CB1R and CB2R are expressed in both the central nervous
creased neuropathic pain in BERK sickle mice (2). The safety system and non-central nervous system tissues, including in-
of sivelestat is already established in humans, and it has been flammatory cells (59, 82). Activation of CB2R peripherally
approved for treatment of acute lung injury in Japan (3). Trials generates an antinociceptive response in inflammatory and
of this drug in the United States were previously halted because neuropathic pain (41). Targeting CB1R has psychotropic ef-
of concerns that it increased long-term mortality rate in me- fects, but targeting CB2R does not have psychotropic effects
chanically ventilated patients with acute lung injury (93) Sive- (27). CB2R agonists and/or knockout mice provide compelling
lestat and other similar drugs in development require preclin- evidence that CB2R activation mitigates neuropathic and in-
ical evaluation for sickle-specific pain and associated adverse flammatory pain and is protective against ischemia-reperfusion
effects. injury by decreasing the endothelial expression of adhesion
molecules and secretion of chemokines (58, 60, 67) and by
Inhibiting Mast Cell Activation attenuating leukocyte adhesion to the endothelium, transendo-
thelial migration, and interrelated oxidative nitrosative damage
Activation of mast cell plays a critical role in neurogenic (63), all of which are consistent with the pathobiology of SCD.
inflammation and nociceptor activation via the release of SP in We showed that cannabinoids mitigate mast cell activation,
the skin and DRG of sickle mice. We found that imatinib, a inflammation, and neurogenic inflammation in BERK sickle
mast cell inhibitor, significantly reduces neurogenic hyperal- mice via both CB1R and CB2R, but activation of CB1R is
gesia and prevents hypoxia-reoxygenation-induced hyperalge- required to ameliorate hyperalgesia (83). In 86 human patients
sia in BERK sickle mice (P ⬍ 0.05; 84). In separate case with HbSS, HbSC, and HbS-␤ thalassemia, a structured self-
reports of two patients with chronic myeloid leukemia and administered anonymous questionnaire found that that 31
SCD, imatinib significantly reduced painful episodes, hospital- (36%) had used cannabis in the previous 12 mo to relieve
izations, and daily pain (21, 74). Imatinib is FDA approved for symptoms associated with SCD (40). The main route in all but
several indications. At this time, we are not aware of any two patients was by smoking. The most common reasons for
clinical trials of imatinib in SCD, but the rationale for such a use were to reduce pain (in 52%) and to induce relaxation or
trial would be compelling. We also found that pretreatment relieve anxiety and depression (in 39%). We are currently
with the mast cell stabilizer cromolyn sodium potentiated the performing a double-blind, placebo-controlled, FDA-approved
analgesic effect of otherwise ineffective low doses of morphine clinical trial to test the effect of vaporized cannabis on chronic
in BERK sickle mice (84). Therefore, it is likely that morphine pain in SCD (ClinicalTrials.gov identifier NCT01771731).
activates mast cells, thereby contributing to pain, but concom-
itantly acts as an analgesic via its activity in the nervous Elevating Fetal Hemoglobin
system. Mast cell targeting thus offers an opioid-reducing
strategy in the management of sickle cell pain. Chronic pain has been found to be inversely associated with
Calpain-1 contributes to IgE-mediated mast cell activation circulating fetal hemoglobin (HbF) in patients with SCD (25).
and plays an important role in neurotransmission and neuro- Notably, higher HbF in patients with SCD has been shown to
genic pain (89). We have demonstrated that genetic ablation of be associated with reduced frequency of painful crises (51).
calpain-1 in Townes sickle mice ameliorates tonic hyperalgesia HbF-increasing strategies have been examined in preclinical
in sickle mice, but not that evoked by hypoxia-reoxygenation studies. The mammalian target of rapamycin (mTOR) plays a
(62). Trifluoperazine, an FDA-approved antipsychotic agent, central role in regulating many fundamental cell processes,
acts as a potent CaMKII␣ inhibitor and reduces calpain-1 from protein synthesis to autophagy, and deregulated mTOR
activity (49). It ameliorated neurogenic pain and caused 50% signaling is implicated in a variety of pathological conditions
reduction in chronic pain in patients with SCD without severe (70). The mTOR inhibitor, rapamycin, increases HbF levels
sedation or supplemental opioid analgesics in a phase I clinical and ameliorates the nociception phenotype in sickle cell mice
trial (57). Statins also decrease calpain-1 activation (91). In a (43). Gaudre et al. reported a case of a kidney transplant
single-center pilot study in adolescents and adults with SCD, recipient with SCD who was treated with everolimus, an
simvastatin treatment for up to 3 mo led to an 85% reduction mTOR inhibitor (33). At 10 mo after initiating therapy, the
in the rate of sickle cell-related pain and oral analgesic use, patient’s HbF level increased dramatically from 4.8 to 15%,
improved soluble biomarkers of inflammation, and acted syn- and there was excellent tolerance to treatment. Reinduction of
ergistically with hydroxyurea (39). Despite the marked decline HbF by depleting DNA methyltransferase 1 with a combina-
in the rate of occurrence of pain, the intensity of pain did not tion of decitabine and tetrahydrouridine led to a significant
change with simvastatin (39). To explain this disparate effect increase in HbF and improved RBC quality as well as hemo-
of simvastatin on frequency and intensity of pain, the authors lysis and inflammation in patients with SCD in a phase I
of this study hypothesized that the visual analog scale, used as clinical trial (NCT01685515). Together, these preclinical and

AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00021.2018 • www.ajpregu.org


Downloaded from journals.physiology.org/journal/ajpregu (103.092.202.027) on August 6, 2023.
TARGETING SICKLE CELL PAIN R109
clinical observations suggest the potential of HbF-enhancing ture-treated mice into three groups on the basis of their increase
strategies in reducing the activation of pain pathways. in pain threshold after electroacupuncture treatment: high re-
sponders (⬎200%), moderate responders (~100 –200%), and
Anti-inflammatory and Antioxidant Strategies nonresponders (ⱕ100%). Electroacupuncture led to reduction
Omega-3 fatty acids have antiaggregatory, antiadhesive, in inflammatory cytokines, SP, and neurogenic inflammation in
anti-inflammatory, and vasodilatory properties. Omega–3 fatty the periphery and signaling pathways of nociception in the
acid was shown to reduce frequency of VOC and transfusion spinal cord and potentiated the effect of a suboptimal dose of
requirements in a randomized study of 140 patients in Sudan morphine in moderate responders, resulting in analgesia equiv-
(23). In the United States, a phase I/II clinical trial to determine alent to that in high responders (85). In a retrospective review
the safety of a new formulation of the omega–3 fatty acids and descriptive analysis of 24 patients with SCD with pain
docosahexaenoic acid and eicosapentaenoic acid and to assess treated at the National Institutes of Health, 9 patients under-
whether it decreases inflammation and inflammatory pain in went only inpatient acupuncture for VOC, 11 patients received
children and young adults with SCD has been undertaken only outpatient acupuncture treatment for chronic pain, and 4
(NCT02947100). patients received both inpatient and outpatient treatments (48).
Central nociceptive mechanisms contribute to chronic pain For the patients who received inpatient acupuncture treatment
and hyperalgesia in SCD, releasing reactive oxidative species, for VOC, there was a significant reduction of reported pain
inflammatory cytokines, neurotrophic factors, and prostaglan- score immediately after acupuncture treatment with an average
dins that excite nociceptive neurons by activated microglia pain reduction of 2.1 points on the 0 –10 numeric pain scale
contributing to the persistence of chronic pain in SCD (34). We (P ⬍ 0.0001). Five of the 15 outpatients had only 1 session per
found that glial activation, neuroinflammation, and oxidative course of acupuncture and thus did not contribute to outpatient
stress in spinal cords of sickle mice are ameliorated with data. Of the 10 outpatients who accepted ⬎1 session per course
curcumin and/or coenzyme Q10 (CoQ10), resulting in attenua- of acupuncture, 2 patients accounted for 79.2% of the 231
tion of hyperalgesia in BERK sickle mice (80). Curcumin and sessions. The investigators therefore analyzed the results from
CoQ10 are likely to be relatively nontoxic adjuvants. In a these two patients separately from the other eight patients. Six
preliminary study in a subpopulation of patients with SCD, out of the remaining eight outpatients described their pain as
CoQ10 treatment reduced inflammation and oxidative stress as improved. The two high-usage patients, who had pain mostly
measured by circulating C-reactive protein and thiobarbituric related to chronic ulcers, reported improved pain compared
acid reactive substances, respectively, and reduced pain during with the prior session in 36.1% of sessions, stable pain in
VOC (75). 28.5%, and worse pain in 11.5%. Data were not available for
23% of the sessions (48). This analysis raises the possibility of
Nociceptin Opioid Receptor Agonists acupuncture as an adjuvant for pain management in SCD (48).
Once pain has been evoked, analgesic strategies are re- Perception-Based Therapies
quired. Opioids exert most of their antinociceptive effect via
the ␮-opioid receptor (MOP/R). In addition to MOP/R, the Nonpharmacological techniques such as guided imagery,
opioid receptor family includes the nociceptin opioid receptor mindfulness, relaxation, hypnosis, and cognitive behavioral
(NOP/R), which is involved in nociceptive signaling via its therapy are effective in alleviating chronic pain in SCD (4, 11,
endogenous peptide ligand nociceptin/orphanin FQ. NOP/Rs 28). Activation of the descending neuromodulatory pathway by
are found in the DRG, spinal cord, and supraspinal regions of nonpharmacologic interventions (e.g., acupuncture, medita-
the brain involved in nociception. We have shown that AT- tion, and yoga) has been shown to reduce peripheral and
200, a high-affinity NOP/R agonist with low efficacy at the central sensitization. In a pilot study of patients with SCD and
MOP/R, ameliorated chronic pain and hypoxia-reoxygenation- healthy controls, a single 30-min hypnosis session decreased
induced mechanical, thermal, and deep tissue/musculoskeletal pain intensity by a moderate amount in patients with SCD (11).
hyperalgesia in HbSS-BERK sickle mice (81). Daily treatment Pain threshold and tolerance increased after hypnosis in the
for 7 days with AT-200 did not cause tolerance and showed a control group, but not in patients with SCD. Patients with SCD
sustained antinociceptive effect, which improved over time and exhibited lower baseline peripheral blood flow and a greater
led to reduced plasma serum amyloid protein, neuropeptides, increase in blood flow after hypnosis than controls, suggesting
inflammatory cytokines, and mast cell activation in the periph- that hypnosis may increase peripheral vasodilation during both
ery. These data suggest that AT-200 ameliorates pain in sickle the anticipation and experience of pain in patients with SCD.
mice via NOP/R by reducing inflammation and mast cell
activation without causing tolerance. Thus, NOP/R agonists are FUTURE PERSPECTIVES
promising drugs for treating pain in SCD because of their Emerging pain therapies are in development that target SCD
effect on sickle pathobiology as well as nociceptive mecha- pathobiology, peripheral and central sensory mechanisms, and
nisms. descending neuromodulatory pathways. Notably, many thera-
Electroacupuncture peutic strategies have the potential to reduce opioid require-
ment and improve opioid analgesia. Targeting pain at its source
We found that electroacupuncture in awake BERK sickle is perhaps the most effective method of preventing the pain of
mice influenced the sickle microenvironment as well as noci- SCD compared with treating pain once it has been evoked.
ception centrally. We observed varied antinociceptive response Targeting mast cells with imatinib, an FDA-approved drug, to
among identical electroacupuncture-treated mice. To analyze ameliorate the evoking of somatosensory mechanisms by
individualized responses, we categorized all electroacupunc- sickle pathobiology appears to be a highly promising pharma-

AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00021.2018 • www.ajpregu.org


Downloaded from journals.physiology.org/journal/ajpregu (103.092.202.027) on August 6, 2023.
R110 TARGETING SICKLE CELL PAIN

cotherapeutic strategy to prevent/treat sickle cell pain. In- Helly M, Gillard E, Sebag G, Kchouk H, Pracros JP, Finck B, Dacher
creased attention is required to apply perception-based affec- JN, Ickowicz V, Raybaud C, Poncet M, Lesprit E, Reinert PH,
Brugières P. Multicenter prospective study of children with sickle cell
tive modulation to minimize the use of analgesics. A major gap disease: radiographic and psychometric correlation. J Child Neurol 15:
still exists in diagnostic criteria for quantifying and phenotyp- 333–343, 2000. doi:10.1177/088307380001500510.
ing pain on the basis of circulating biomarkers, physiological 11. Bhatt RR, Martin SR, Evans S, Lung K, Coates TD, Zeltzer LK, Tsao
measures, and neuroimaging to guide the use of specific JC. The effect of hypnosis on pain and peripheral blood flow in sickle-cell
therapies and/or combination therapies. Finally, well-designed disease: a pilot study. J Pain Res 10: 1635–1644, 2017. doi:10.2147/JPR.
S131859.
clinical trials are required to test these pharmacological and 12. Brandow AM, Stucky CL, Hillery CA, Hoffmann RG, Panepinto JA.
integrative approaches. Patients with sickle cell disease have increased sensitivity to cold and heat.
Am J Hematol 88: 37–43, 2013. doi:10.1002/ajh.23341.
ACKNOWLEDGMENTS 13. Brown RT, Davis PC, Lambert R, Hsu L, Hopkins K, Eckman J.
We thank Michael Franklin for editorial suggestions and Barb Benson for Neurocognitive functioning and magnetic resonance imaging in children
assistance with manuscript preparation. We apologize for our inability to with sickle cell disease. J Pediatr Psychol 25: 503–513, 2000. doi:10.
include many studies due to space limitations, but we deeply appreciate their 1093/jpepsy/25.7.503.
contribution to the study of pain/SCD. 14. Buchanan G, Yawn B, Afenyi-Annan A, Ballas S, Hassell K, James A,
Jordan L, Lanzkron S, Lottenberg R, Savage W, Tanabe P, Ware RE.
GRANTS Evidence-Based Management of Sickle Cell Disease: Expert Panel Report,
2014. Bethesda, MD: National Heart, Lung, and Blood Institute, 2014.
This effort was supported by National Heart, Lung, and Blood Institute https://www.nhlbi.nih.gov/health-topics/evidence-based-management-
Grant U01-HL-117664 to Kalpna Gupta. sickle-cell-disease.
15. Buckner RL, Andrews-Hanna JR, Schacter DL. The brain’s default
DISCLAIMERS network: anatomy, function, and relevance to disease. Ann NY Acad Sci
The content is solely the responsibility of the authors and does not 1124: 1–38, 2008. doi:10.1196/annals.1440.011.
necessarily represent the official views of the National Institutes of Health. 16. Bunn HF. Pathogenesis and treatment of sickle cell disease. N Engl J Med
337: 762–769, 1997. doi:10.1056/NEJM199709113371107.
DISCLOSURES 17. Cain DM, Vang D, Simone DA, Hebbel RP, Gupta K. Mouse models
for studying pain in sickle disease: effects of strain, age, and acuteness. Br
Kalpna Gupta is a consultant for TauTona Group. J Haematol 156: 535–544, 2012. doi:10.1111/j.1365-2141.2011.08977.x.
18. Campbell CM, Moscou-Jackson G, Carroll CP, Kiley K, Haywood C
AUTHOR CONTRIBUTIONS
Jr, Lanzkron S, Hand M, Edwards RR, Haythornthwaite JA. An
K.G. and K.G. prepared figures; K.G., O.J., and K.G. drafted manuscript; evaluation of central sensitization in patients with sickle cell disease. J
K.G., O.J., and K.G. edited and revised manuscript; K.G., O.J., and K.G. Pain 17: 617–627, 2016. doi:10.1016/j.jpain.2016.01.475.
approved final version of manuscript; Kalpna G. conceived and designed 19. Case M, Zhang H, Mundahl J, Datta Y, Nelson S, Gupta K, He B.
research. Characterization of functional brain activity and connectivity using EEG
and fMRI in patients with sickle cell disease. Neuroimage Clin 14: 1–17,
REFERNCES 2017. doi:10.1016/j.nicl.2016.12.024.
20. Cataldo G, Rajput S, Gupta K, Simone DA. Sensitization of nociceptive
1. Aich A, Beitz AJ, Gupta K. Mechanisms of pain in sickle cell disease. In: spinal neurons contributes to pain in a transgenic model of sickle cell
Sickle Cell Disease: Pain and Common Chronic Complications, edited by disease. Pain 156: 722–730, 2015. doi:10.1097/j.pain.0000000000000104.
Inusa BP. London: InTech, 2016, chapt. 5, p. 69 –91. doi:10.5772/64647. 21. Close JL, Lottenberg R. Effectiveness of imatinib therapy for a patient
https://www.intechopen.com/books/sickle-cell-disease-pain-and-common- with sickle cell anemia and chronic myelocytic leukemia. Blood 114:
chronic-complications/mechanisms-of-pain-in-sickle-cell-disease. 2559, 2009.
2. Aich A, Paul J, Lei J, Wang Y, Bagchi A, Gupta K. Regulation of 22. Cohen MJ, Branch WB, McKie VC, Adams RJ. Neuropsychological
elastase by SerpinA3N contributes to pain in sickle cell disease. Blood
impairment in children with sickle cell anemia and cerebrovascular
128: 858, 2016.
accidents. Clin Pediatr (Phila) 33: 517–524, 1994. doi:10.1177/
3. Aikawa N, Kawasaki Y. Clinical utility of the neutrophil elastase inhib-
000992289403300902.
itor sivelestat for the treatment of acute respiratory distress syndrome.
23. Daak AA, Ghebremeskel K, Hassan Z, Attallah B, Azan HH, Elbashir
Ther Clin Risk Manag 10: 621–629, 2014. doi:10.2147/TCRM.S65066.
MI, Crawford M. Effect of omega–3 (n-3) fatty acid supplementation in
4. Apkarian AV, Bushnell MC, Treede RD, Zubieta JK. Human brain
patients with sickle cell anemia: randomized, double-blind, placebo-
mechanisms of pain perception and regulation in health and disease. Eur
J Pain 9: 463–484, 2005. doi:10.1016/j.ejpain.2004.11.001. controlled trial. Am J Clin Nutr 97: 37–44, 2013. doi:10.3945/ajcn.112.
5. Armstrong FD, Thompson RJ Jr, Wang W, Zimmerman R, Pegelow 036319.
CH, Miller S, Moser F, Bello J, Hurtig A, Vass K; Neuropsychology 24. Dampier C, Palermo TM, Darbari DS, Hassell K, Smith W, Zempsky
Committee of the Cooperative Study of Sickle Cell Disease. Cognitive W. AAPT diagnostic criteria for chronic sickle cell disease pain. J Pain
functioning and brain magnetic resonance imaging in children with sickle 18: 490 –498, 2017. doi:10.1016/j.jpain.2016.12.016.
cell disease. Pediatrics 97: 864 –870, 1996. 25. Darbari DS, Hampson JP, Ichesco E, Kadom N, Vezina G, Evangelou
6. Ataga KI, Kutlar A, Kanter J, Liles D, Cancado R, Friedrisch J, I, Clauw DJ, Taylor Vi JG, Harris RE. Frequency of hospitalizations for
Guthrie TH, Knight-Madden J, Alvarez OA, Gordeuk VR, Gua- pain and association with altered brain network connectivity in sickle cell
landro S, Colella MP, Smith WR, Rollins SA, Stocker JW, Rother RP. disease. J Pain 16: 1077–1086, 2015. doi:10.1016/j.jpain.2015.07.005.
Crizanlizumab for the prevention of pain crises in sickle cell disease. N 26. DeBaun MR, Schatz J, Siegel MJ, Koby M, Craft S, Resar L, Chu JY,
Engl J Med 376: 429 –439, 2017. doi:10.1056/NEJMoa1611770. Launius G, Dadash-Zadeh M, Lee RB, Noetzel M. Cognitive screening
7. Ballas SK. Sickle Cell Pain. Philadelphia, PA: Lippincott Williams & examinations for silent cerebral infarcts in sickle cell disease. Neurology
Wilkins, 2015. 50: 1678 –1682, 1998. doi:10.1212/WNL.50.6.1678.
8. Ballas SK, Gupta K, Adams-Graves P. Sickle cell pain: a critical 27. Desroches J, Bouchard JF, Gendron L, Beaulieu P. Involvement of
reappraisal. Blood 120: 3647–3656, 2012. doi:10.1182/blood-2012-04- cannabinoid receptors in peripheral and spinal morphine analgesia. Neu-
383430. roscience 261: 23–42, 2014. doi:10.1016/j.neuroscience.2013.12.030.
9. Beckmann CF, DeLuca M, Devlin JT, Smith SM. Investigations into 28. Evans S, Seidman L, Sternlieb B, Casillas J, Zeltzer L, Tsao J. Clinical
resting-state connectivity using independent component analysis. Philos case report: yoga for fatigue in five young adult survivors of childhood
Trans R Soc Lond B Biol Sci 360: 1001–1013, 2005. doi:10.1098/rstb. cancer. J Adolesc Young Adult Oncol 6: 96 –101, 2017. doi:10.1089/jayao.
2005.1634. 2016.0013.
10. Bernaudin F, Verlhac S, Fréard F, Roudot-Thoraval F, Benkerrou M, 29. Fabry ME, Nagel RL, Pachnis A, Suzuka SM, Costantini F. High
Thuret I, Mardini R, Vannier JP, Ploix E, Romero M, Cassé-Perrot C, expression of human beta S- and alpha-globins in transgenic mice:

AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00021.2018 • www.ajpregu.org


Downloaded from journals.physiology.org/journal/ajpregu (103.092.202.027) on August 6, 2023.
TARGETING SICKLE CELL PAIN R111
hemoglobin composition and hematological consequences. Proc Natl 48. Lu K, Cheng MC, Ge X, Berger A, Xu D, Kato GJ, Minniti CP. A
Acad Sci USA 89: 12150 –12154, 1992. doi:10.1073/pnas.89.24.12150. retrospective review of acupuncture use for the treatment of pain in sickle
30. Fabry ME, Sengupta A, Suzuka SM, Costantini F, Rubin EM, Ho- cell disease patients: descriptive analysis from a single institution. Clin J
frichter J, Christoph G, Manci E, Culberson D, Factor SM, Nagel RL. Pain 30: 825–830, 2014. doi:10.1097/AJP.0000000000000036.
A second generation transgenic mouse model expressing both hemoglobin 49. Luo F, Yang C, Chen Y, Shukla P, Tang L, Wang LX, Wang ZJ.
S (HbS) and HbS-Antilles results in increased phenotypic severity. Blood Reversal of chronic inflammatory pain by acute inhibition of Ca2⫹/
86: 2419 –2428, 1995. calmodulin-dependent protein kinase II. J Pharmacol Exp Ther 325:
31. Finan PH, Carroll CP, Moscou-Jackson G, Martel MO, Campbell 267–275, 2008. doi:10.1124/jpet.107.132167.
CM, Pressman A, Smyth JM, Tremblay JM, Lanzkron SM, Hay- 50. Manwani D, Frenette PS. Vaso-occlusion in sickle cell disease: patho-
thornthwaite JA. Daily opioid use fluctuates as a function of pain, physiology and novel targeted therapies. Blood 122: 3892–3898, 2013.
catastrophizing, and affect in patients with sickle cell disease: an elec- doi:10.1182/blood-2013-05-498311.
tronic daily diary analysis. J Pain 19: 46 –56, 2018. doi:10.1016/j.jpain. 51. Mashon RS, Dash PM, Khalkho J, Dash L, Mohanty PK, Patel S,
2017.08.010. Mohanty RC, Das BS, Das UK, Das PK, Patel DK. Higher fetal
32. Gaboury JP, Johnston B, Niu XF, Kubes P. Mechanisms underlying hemoglobin concentration in patients with sickle cell disease in eastern
acute mast cell-induced leukocyte rolling and adhesion in vivo. J Immunol India reduces frequency of painful crisis. Eur J Haematol 83: 383–384,
154: 804 –813, 1995. 2009. doi:10.1111/j.1600-0609.2009.01290.x.
33. Gaudre N, Cougoul P, Bartolucci P, Dörr G, Bura-Riviere A, Kamar 52. Matsui NM, Borsig L, Rosen SD, Yaghmai M, Varki A, Embury SH.
N, Del Bello A. Improved fetal hemoglobin with mTOR inhibitor-based P-selectin mediates the adhesion of sickle erythrocytes to the endothelium.
immunosuppression in a kidney transplant recipient with sickle cell Blood 98: 1955–1962, 2001. doi:10.1182/blood.V98.6.1955.
disease. Am J Transplant 17: 2212–2214, 2017. doi:10.1111/ajt.14263. 53. Mayer EA, Labus JS, Tillisch K, Cole SW, Baldi P. Towards a systems
34. Grace PM, Hutchinson MR, Maier SF, Watkins LR. Pathological pain view of IBS. Nat Rev Gastroenterol Hepatol 12: 592–605, 2015. doi:10.
and the neuroimmune interface. Nat Rev Immunol 14: 217–231, 2014. 1038/nrgastro.2015.121.
doi:10.1038/nri3621. 54. McClish DK, Smith WR, Levenson JL, Aisiku IP, Roberts JD, Roseff
35. Gupta K. Iatrogenic angiogenesis. In: Morphine and Metastasis, edited by SD, Bovbjerg VE. Comorbidity, pain, utilization, and psychosocial out-
Parat MO. Dordrecht, The Netherlands: Springer, 2013, p. 63–78. doi:10. comes in older versus younger sickle cell adults: the PiSCES project.
1007/978-94-007-5678-6_5. BioMed Res Int 2017: 4070547, 2017. doi:10.1155/2017/4070547.
36. Gupta K, Harvima IT. Mast cell-neural interactions contribute to pain 55. Michaels LA, Ohene-Frempong K, Zhao H, Douglas SD. Serum levels
and itch. Immunol Rev 282: 168 –187, 2018. doi:10.1111/imr.12622. of substance P are elevated in patients with sickle cell disease and increase
37. Gupta M, Msambichaka L, Ballas SK, Gupta K. Morphine for the further during vaso-occlusive crisis. Blood 92: 3148 –3151, 1998.
treatment of pain in sickle cell disease. Sci World J 2015: 540154, 2015. 56. Mittal A, Gupta M, Lamarre Y, Jahagirdar B, Gupta K. Quantification
doi:10.1155/2015/540154. of pain in sickle mice using facial expressions and body measurements.
38. Hillery CA, Kerstein PC, Vilceanu D, Barabas ME, Retherford D, Blood Cells Mol Dis 57: 58 –66, 2016. doi:10.1016/j.bcmd.2015.12.006.
Brandow AM, Wandersee NJ, Stucky CL. Transient receptor potential 57. Molokie RE, Wilkie DJ, Wittert H, Suarez ML, Yao Y, Zhao Z, He Y,
vanilloid 1 mediates pain in mice with severe sickle cell disease. Blood Wang ZJ. Mechanism-driven phase I translational study of trifluoperazine
118: 3376 –3383, 2011. doi:10.1182/blood-2010-12-327429. in adults with sickle cell disease. Eur J Pharmacol 723: 419 –424, 2014.
39. Hoppe C, Jacob E, Styles L, Kuypers F, Larkin S, Vichinsky E. doi:10.1016/j.ejphar.2013.10.062.
Simvastatin reduces vaso-occlusive pain in sickle cell anaemia: a pilot 58. Montecucco F, Lenglet S, Braunersreuther V, Burger F, Pelli G,
efficacy trial. Br J Haematol 177: 620 –629, 2017. doi:10.1111/bjh.14580. Bertolotto M, Mach F, Steffens S. CB2 cannabinoid receptor activation
40. Howard J, Anie KA, Holdcroft A, Korn S, Davies SC. Cannabis use in is cardioprotective in a mouse model of ischemia/reperfusion. J Mol Cell
sickle cell disease: a questionnaire study. Br J Haematol 131: 123–128, Cardiol 46: 612–620, 2009. doi:10.1016/j.yjmcc.2008.12.014.
2005. doi:10.1111/j.1365-2141.2005.05723.x. 59. Munro S, Thomas KL, Abu-Shaar M. Molecular characterization of a
41. Ibrahim MM, Deng H, Zvonok A, Cockayne DA, Kwan J, Mata HP, peripheral receptor for cannabinoids. Nature 365: 61–65, 1993. doi:10.
Vanderah TW, Lai J, Porreca F, Makriyannis A, Malan TP Jr. 1038/365061a0.
Activation of CB2 cannabinoid receptors by AM1241 inhibits experimen- 60. Murikinati S, Jüttler E, Keinert T, Ridder DA, Muhammad S,
tal neuropathic pain: pain inhibition by receptors not present in the CNS. Waibler Z, Ledent C, Zimmer A, Kalinke U, Schwaninger M. Acti-
Proc Natl Acad Sci USA 100: 10529 –10533, 2003. doi:10.1073/pnas. vation of cannabinoid 2 receptors protects against cerebral ischemia by
1834309100. inhibiting neutrophil recruitment. FASEB J 24: 788 –798, 2010. doi:10.
42. Jacob E, Chan VW, Hodge C, Zeltzer L, Zurakowski D, Sethna NF. 1096/fj.09-141275.
Sensory and thermal quantitative testing in children with sickle cell 61. Murphy M, Close J, Lottenberg R, Rajasekhar A. Effectiveness of
disease. J Pediatr Hematol Oncol 37: 185–189, 2015. doi:10.1097/MPH. imatinib therapy for sickle cell anemia and chronic myeloid leukemia. Am
0000000000000214. J Med Sci 347: 254 –255, 2014. doi:10.1097/MAJ.0000000000000228.
43. Khaibullina A, Almeida LE, Wang L, Kamimura S, Wong EC, 62. Nwankwo JO, Lei J, Xu J, Rivera A, Gupta K, Chishti AH. Genetic
Nouraie M, Maric I, Albani S, Finkel J, Quezado ZM. Rapamycin inactivation of calpain-1 attenuates pain sensitivity in a humanized mouse
increases fetal hemoglobin and ameliorates the nociception phenotype in model of sickle cell disease. Haematologica 101: e397–e400, 2016.
sickle cell mice. Blood Cells Mol Dis 55: 363–372, 2015. doi:10.1016/j. doi:10.3324/haematol.2016.148106.
bcmd.2015.08.001. 63. Pacher P, Haskó G. Endocannabinoids and cannabinoid receptors in
44. Klein LM, Lavker RM, Matis WL, Murphy GF. Degranulation of ischaemia-reperfusion injury and preconditioning. Br J Pharmacol 153:
human mast cells induces an endothelial antigen central to leukocyte 252–262, 2008. doi:10.1038/sj.bjp.0707582.
adhesion. Proc Natl Acad Sci USA 86: 8972–8976, 1989. doi:10.1073/ 64. Pászty C, Brion CM, Manci E, Witkowska HE, Stevens ME, Mohan-
pnas.86.22.8972. das N, Rubin EM. Transgenic knockout mice with exclusively human
45. Kohli DR, Li Y, Khasabov SG, Gupta P, Kehl LJ, Ericson ME, sickle hemoglobin and sickle cell disease. Science 278: 876 –878, 1997.
Nguyen J, Gupta V, Hebbel RP, Simone DA, Gupta K. Pain-related doi:10.1126/science.278.5339.876.
behaviors and neurochemical alterations in mice expressing sickle hemo- 65. Piel FB, Steinberg MH, Rees DC. Sickle cell disease. N Engl J Med 376:
globin: modulation by cannabinoids. Blood 116: 456 –465, 2010. doi:10. 1561–1573, 2017. doi:10.1056/NEJMra1510865.
1182/blood-2010-01-260372. 66. Polanowska-Grabowska R, Wallace K, Field JJ, Chen L, Marshall
46. Kutlar A, Ataga KI, McMahon L, Howard J, Galacteros F, Hagar W, MA, Figler R, Gear AR, Linden J. P-selectin-mediated platelet-neutro-
Vichinsky E, Cheung AT, Matsui N, Embury SH. A potent oral phil aggregate formation activates neutrophils in mouse and human sickle
P-selectin blocking agent improves microcirculatory blood flow and a cell disease. Arterioscler Thromb Vasc Biol 30: 2392–2399, 2010. doi:10.
marker of endothelial cell injury in patients with sickle cell disease. Am J 1161/ATVBAHA.110.211615.
Hematol 87: 536 –539, 2012. doi:10.1002/ajh.23147. 67. Rajesh M, Pan H, Mukhopadhyay P, Bátkai S, Osei-Hyiaman D,
47. Lei J, Benson B, Tran H, Ofori-Acquah SF, Gupta K. Comparative Haskó G, Liaudet L, Gao B, Pacher P. Cannabinoid-2 receptor agonist
analysis of pain behaviours in humanized mouse models of sickle cell HU-308 protects against hepatic ischemia/reperfusion injury by attenuat-
anemia. PLoS One 11: e0160608, 2016. doi:10.1371/journal.pone. ing oxidative stress, inflammatory response, and apoptosis. J Leukoc Biol
0160608. 82: 1382–1389, 2007. doi:10.1189/jlb.0307180.

AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00021.2018 • www.ajpregu.org


Downloaded from journals.physiology.org/journal/ajpregu (103.092.202.027) on August 6, 2023.
R112 TARGETING SICKLE CELL PAIN

68. Rees DC, Williams TN, Gladwin MT. Sickle-cell disease. Lancet 376: 82. Van Sickle MD, Duncan M, Kingsley PJ, Mouihate A, Urbani P,
2018 –2031, 2010. doi:10.1016/S0140-6736(10)61029-X. Mackie K, Stella N, Makriyannis A, Piomelli D, Davison JS, Marnett
69. Ryan TM, Ciavatta DJ, Townes TM. Knockout-transgenic mouse model LJ, Di Marzo V, Pittman QJ, Patel KD, Sharkey KA. Identification and
of sickle cell disease. Science 278: 873–876, 1997. doi:10.1126/science. functional characterization of brainstem cannabinoid CB2 receptors. Sci-
278.5339.873. ence 310: 329 –332, 2005. doi:10.1126/science.1115740.
70. Saxton RA, Sabatini DM. mTOR signaling in growth, metabolism, and 82a.Vicuña L, Strochlic DE, Latremoliere A, Bali KK, Simonetti M,
disease. Cell 168: 960 –976, 2017. [Erratum in Cell 169: 361–371, 2017. Husainie D, Prokosch S, Riva P, Griffin RS, Njoo C, Gehrig S, Mall
10.1016/j.cell.2017.03.035.] doi:10.1016/j.cell.2017.02.004. MA, Arnold B, Devor M, Woolf CJ, Liberles SD, Costigan M, Kuner
71. Schatz J, Buzan R. Decreased corpus callosum size in sickle cell disease: R, The serine protease inhibitor SerpinA3N attenuates neuropathic pain by
relationship with cerebral infarcts and cognitive functioning. J Int Neuro- inhibiting T cell-derived leukocyte elastase. Nat Med 21: 518 –523, 2015.
psychol Soc 12: 24 –33, 2006. doi:10.1017/S1355617706060085. doi:10.1038/nm.3852.
72. Small-Howard AL, Shimoda LM, Adra CN, Turner H. Anti-inflam- 83. Vincent L, Vang D, Nguyen J, Benson B, Lei J, Gupta K. Cannabinoid
matory potential of CB1-mediated cAMP elevation in mast cells. Biochem receptor-specific mechanisms to alleviate pain in sickle cell anemia via
J 388: 465–473, 2005. doi:10.1042/BJ20041682. inhibition of mast cell activation and neurogenic inflammation. Haema-
73. Smith WR, Bauserman RL, Ballas SK, McCarthy WF, Steinberg MH, tologica 101: 566 –577, 2016. doi:10.3324/haematol.2015.136523.
Swerdlow PS, Waclawiw MA, Barton BA; Multicenter Study of 84. Vincent L, Vang D, Nguyen J, Gupta M, Luk K, Ericson ME, Simone
Hydroxyurea in Sickle Cell Anemia. Climatic and geographic temporal DA, Gupta K. Mast cell activation contributes to sickle cell pathobiology
patterns of pain in the Multicenter Study of Hydroxyurea. Pain 146: and pain in mice. Blood 122: 1853–1862, 2013. doi:10.1182/blood-2013-
91–98, 2009. doi:10.1016/j.pain.2009.07.008. 04-498105.
74. Stankovic Stojanovic K, Thiolière B, Garandeau E, Lecomte I, Bach- 85. Wang Y, Lei J, Gupta M, Peng F, Lam S, Jha R, Raduenz E, Beitz AJ,
meyer C, Lionnet F. Chronic myeloid leukaemia and sickle cell disease: Gupta K. Electroacupuncture in conscious free-moving mice reduces pain
could imatinib prevent vaso-occlusive crisis? Br J Haematol 155: 271– by ameliorating peripheral and central nociceptive mechanisms. Sci Rep 6:
34493, 2016. doi:10.1038/srep34493.
272, 2011. doi:10.1111/j.1365-2141.2011.08670.x.
86. Ware RE, de Montalembert M, Tshilolo L, Abboud MR. Sickle cell
75. Thakur AS, Littaru GP, Moesgaard S, Dan Sindberg C, Khan Y,
disease. Lancet 390: 311–323, 2017. doi:10.1016/S0140-6736(17)30193-9.
Singh CM. Hematological parameters and RBC TBARS level of Q 10
87. Watkins KE, Hewes DK, Connelly A, Kendall BE, Kingsley DP, Evans
supplemented tribal sickle cell patients: a hospital based study. Indian J
JE, Gadian DG, Vargha-Khadem F, Kirkham FJ. Cognitive deficits
Clin Biochem 28: 185–188, 2013. doi:10.1007/s12291-012-0277-9.
associated with frontal-lobe infarction in children with sickle cell disease.
76. Thorlacius H, Raud J, Rosengren-Beezley S, Forrest MJ, Hedqvist P,
Dev Med Child Neurol 40: 536 –543, 1998. doi:10.1111/j.1469-8749.
Lindbom L. Mast cell activation induces P-selectin-dependent leukocyte 1998.tb15412.x.
rolling and adhesion in postcapillary venules in vivo. Biochem Biophys 88. Weyer AD, Stucky CL. Repurposing a leukocyte elastase inhibitor for
Res Commun 203: 1043–1049, 1994. doi:10.1006/bbrc.1994.2287. neuropathic pain. Nat Med 21: 429 –430, 2015. doi:10.1038/nm.3861.
77. Tran H, Gupta M, Gupta K. Targeting novel mechanisms of pain in 89. Wu Z, Chen X, Liu F, Chen W, Wu P, Wieschhaus AJ, Chishti AH,
sickle cell disease. Blood 130: 2377–2385, 2017. doi:10.1182/blood-2017- Roche PA, Chen WM, Lin TJ. Calpain-1 contributes to IgE-mediated
05-782003. mast cell activation. J Immunol 192: 5130 –5139, 2014. doi:10.4049/
78. Turhan A, Weiss LA, Mohandas N, Coller BS, Frenette PS. Primary jimmunol.1301677.
role for adherent leukocytes in sickle cell vascular occlusion: a new 90. Wun T, Paglieroni T, Tablin F, Welborn J, Nelson K, Cheung A.
paradigm. Proc Natl Acad Sci USA 99: 3047–3051, 2002. doi:10.1073/ Platelet activation and platelet-erythrocyte aggregates in patients with
pnas.052522799. sickle cell anemia. J Lab Clin Med 129: 507–516, 1997. doi:10.1016/
79. Uhelski ML, Gupta K, Simone DA. Sensitization of C-fiber nociceptors S0022-2143(97)90005-6.
in mice with sickle cell disease is decreased by local inhibition of 91. Yin M, Liu Q, Yu L, Yang Y, Lu M, Wang H, Luo D, Rong X, Tang
anandamide hydrolysis. Pain 158: 1711–1722, 2017. doi:10.1097/j.pain. F, Guo J. Downregulations of CD36 and calpain-1, inflammation, and
0000000000000966. atherosclerosis by simvastatin in apolipoprotein E knockout mice. J Vasc
80. Valverde Y, Benson B, Gupta M, Gupta K. Spinal glial activation and Res 54: 123–130, 2017. doi:10.1159/000464288.
oxidative stress are alleviated by treatment with curcumin or coenzyme Q 92. Zarbock A, Polanowska-Grabowska RK, Ley K. Platelet-neutrophil-
in sickle mice. Haematologica 101: e44 –e47, 2016. doi:10.3324/ interactions: linking hemostasis and inflammation. Blood Rev 21: 99 –111,
haematol.2015.137489. 2007. doi:10.1016/j.blre.2006.06.001.
81. Vang D, Paul JA, Nguyen J, Tran H, Vincent L, Yasuda D, Zaveri NT, 93. Zeiher BG, Artigas A, Vincent JL, Dmitrienko A, Jackson K, Thomp-
Gupta K. Small-molecule nociceptin receptor agonist ameliorates mast son BT, Bernard G; STRIVE Study Group. Neutrophil elastase inhi-
cell activation and pain in sickle mice. Haematologica 100: 1517–1525, bition in acute lung injury: results of the STRIVE study. Crit Care Med
2015. doi:10.3324/haematol.2015.128736. 32: 1695–1702, 2004. doi:10.1097/01.CCM.0000133332.48386.85.

AJP-Regul Integr Comp Physiol • doi:10.1152/ajpregu.00021.2018 • www.ajpregu.org


Downloaded from journals.physiology.org/journal/ajpregu (103.092.202.027) on August 6, 2023.

You might also like